EP3121277B1 - Zur spezifischen bindung an ab-oligomer fähiger antikörper und verwendung davon - Google Patents

Zur spezifischen bindung an ab-oligomer fähiger antikörper und verwendung davon Download PDF

Info

Publication number
EP3121277B1
EP3121277B1 EP16183829.7A EP16183829A EP3121277B1 EP 3121277 B1 EP3121277 B1 EP 3121277B1 EP 16183829 A EP16183829 A EP 16183829A EP 3121277 B1 EP3121277 B1 EP 3121277B1
Authority
EP
European Patent Office
Prior art keywords
antibody
seq
amino acid
acid sequence
chain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
EP16183829.7A
Other languages
English (en)
French (fr)
Other versions
EP3121277A1 (de
Inventor
Etsuro Matsubara
Masao Shibata
Tatsuki Yokoseki
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
National Center for Geriatrics and Gerontology
Immunas Pharma Inc
Original Assignee
National Center for Geriatrics and Gerontology
Immunas Pharma Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by National Center for Geriatrics and Gerontology, Immunas Pharma Inc filed Critical National Center for Geriatrics and Gerontology
Publication of EP3121277A1 publication Critical patent/EP3121277A1/de
Application granted granted Critical
Publication of EP3121277B1 publication Critical patent/EP3121277B1/de
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/68Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids
    • G01N33/6893Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing involving proteins, peptides or amino acids related to diseases not provided for elsewhere
    • G01N33/6896Neurological disorders, e.g. Alzheimer's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/435Assays involving biological materials from specific organisms or of a specific nature from animals; from humans
    • G01N2333/46Assays involving biological materials from specific organisms or of a specific nature from animals; from humans from vertebrates
    • G01N2333/47Assays involving proteins of known structure or function as defined in the subgroups
    • G01N2333/4701Details
    • G01N2333/4709Amyloid plaque core protein
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2800/00Detection or diagnosis of diseases
    • G01N2800/28Neurological disorders
    • G01N2800/2814Dementia; Cognitive disorders
    • G01N2800/2821Alzheimer

Definitions

  • the present invention relates to antibodies that specifically bind to A ⁇ oligomers and uses thereof.
  • Non-Patent Documents 1 and 2 Various evidence has shown that deterioration of memory arises from synaptic dysfunction triggered by soluble A ⁇ oligomers (see Non-Patent Documents 1 and 2). Excessive accumulation and deposition of A ⁇ oligomers may be the trigger for a series of pathological cascades that lead to Alzheimer's disease (AD). Therefore, therapeutic intervention targeting A ⁇ oligomers may be effective for blocking these cascades.
  • findings on neurodegeneration mediated by core molecules of this amyloid cascade hypothesis which are responsible for neurodegeneration, particularly by A ⁇ oligomers originate from in vitro experiments (see Non-Patent Document 3). This neurodegeneration has not been proven directly in vivo.
  • Non-Patent Document 4 There has been known no technique capable of proving the toxicity within the human brain, an aspect which is difficult to demonstrate even in Alzheimer's disease mouse models. Thus, the in vivo neurotoxicity of endogenous A ⁇ has been often disregarded. It has been unknown why NFT formation and loss of nerve cells precede senile plaque formation in the human entorhinal cortex, and how A ⁇ oligomers are involved in this mechanism.
  • An objective of the present invention is to provide antibodies that bind specifically to A ⁇ oligomers, but not to an A ⁇ monomer, and uses thereof. More specifically, the present invention provides an antibody that binds specifically to an A ⁇ oligomer but not to an A ⁇ monomer, wherein the antibody is a monoclonal antibody and is selected from the group consisting of:
  • the present inventors produced monoclonal antibodies that are specific to only soluble amyloid ⁇ (A ⁇ ) oligomers and do not recognize soluble A ⁇ monomers which are physiological molecules, and confirmed that the antibodies have the following:
  • monoclonal 1A9 and 2C3 were determined to specifically recognize oligomers of 30 kDa or more, mainly 100 kDa or more, but not monomers of approximately 4.5 kDa.
  • the two antibodies were confirmed to have neurotoxicity-neutralizing activity by evaluating the neutralizing effect against A ⁇ 1-42-induced neurotoxicity in PC12 cells differentiated into nerve cells.
  • Thioflavin T assay and electron microscopy showed that the antibodies have activity to suppress A ⁇ amyloid fibril formation.
  • the ability of 1A9 and 2C3 to capture A ⁇ oligomers in AD brain was confirmed by immunoprecipitation using the antibodies in the presence of SDS-stable 4-, 5-, 8-, and 12-mers.
  • the amount of polymers recognized by the antibodies was evaluated in the human entorhinal cortex mostly at Braak NFT Stages I to III.
  • the 12-mer which has been reported to have neurotoxicity in animal studies, it was confirmed that the polymer accumulation precedes the occurrence of cognitive impairment, and is increased with the progression of Braak NFT stage.
  • This result shows for the first time that the 12-mer, which is specifically recognized by the antibodies, is a conformational assembly that causes in vivo neurotoxicity in the human brain.
  • the present inventors also discovered that the oligomeric conformational structure recognized by the antibodies is present in cerebrospinal fluid (CSF), and is increased in AD patients.
  • CSF cerebrospinal fluid
  • the present inventors used 1A9 or 2C3 in passive immunotherapy by intravenous injection as with other neurological disorders. It was confirmed that Tg2576 mice are protected from memory impairment, senile plaque formation, synaptic dysfunction, and A ⁇ accumulation by subchronic passive immunotherapy, without harmful side-effects. The results obtained by the present inventors demonstrated for the first time that monoclonal 1A9 and 2C3 are promising candidates for therapeutic antibodies for preventing Alzheimer's disease-like phenotypes in Tg2576 mice, which are expected to show their effect by conventional peripheral intravenous administration, and thus there is no need to consider brain transfer.
  • the present inventors also confirmed that passive immunotherapy using the 1A9 and 2C3 antibodies suppresses senile plaque amyloid formation and swollen dystrophic neurite formation. Furthermore, the present inventors discovered that a fraction of the 1A9 and 2C3 antibodies administered into the blood transfers into the brain.
  • monoclonal 1A9 and 2C3 which are antibodies that specifically bind to A ⁇ oligomers, fulfill all of the diagnostic/therapeutic antibody criteria, and are promising candidates for therapeutic antibodies for diagnosing/preventing Alzheimer's disease.
  • the present inventors successfully obtained the 5A5, 5A9, 4F7, 4H5, 6E4, and 6H4 antibodies which bind specifically to A ⁇ oligomers, but do not recognize A ⁇ monomers.
  • the present inventors discovered that these six types of antibodies have activity to neutralize A ⁇ -induced neurotoxicity and to suppress A ⁇ amyloid fibril formation.
  • the present inventors disclose that the above-mentioned 5A5, 5A9, 4F7, 4H5, 6E4, and 6H4 antibodies are promising candidates for therapeutic antibodies for diagnosing/preventing Alzheimer's disease.
  • the antibodies provided by the present invention are expected to greatly contribute to the establishment of preventive/therapeutic methods selective to molecules responsible for evoking pathological conditions of Alzheimer's disease, and the establishment of early diagnostic markers for Alzheimer's disease.
  • the present inventors obtained evidence showing that, even in antibody therapy targeting pathological conditions in the brain, peripheral intravenous administration is sufficient and there is no need to consider brain transfer.
  • the present invention is expected to rapidly accelerate the progress of antibody drugs for Alzheimer's disease.
  • the present inventors succeeded in obtaining antibodies that bind specifically to A ⁇ oligomers but not to A ⁇ monomers. That is, the present invention provides antibodies that bind to A ⁇ oligomers but not to A ⁇ monomers.
  • the antibodies are preferably isolated or purified.
  • isolated and purified antibodies used for substances (antibodies and such) of the present invention indicate that the substances do not substantially include at least one substance that may be contained in the natural source. Therefore, “isolated antibodies” and “purified antibodies” refer to antibodies that do not substantially include cell materials such as hydrocarbons, lipids, or other contaminant proteins from the cell or tissue source from which the antibodies (proteins) are derived. When the antibodies are chemically synthesized, the terms refer to antibodies that do not substantially include chemical precursor substances or other chemical substances. In a preferred embodiment, the antibodies of the present invention are isolated or purified.
  • Antibodies refers to glycoproteins that have the same structural characteristics. Antibodies show binding specificity towards specific antigens.
  • antigens refers to proteins that have the ability to bind to the corresponding antibodies, and induce antigen-antibody reactions in vivo.
  • a ⁇ proteins which are the major constituents of amyloids, are peptides consisting of 40 to 42 amino acids, and are known to be produced from precursor proteins called amyloid precursor proteins (APPs) by the action of proteases. Besides amyloid fibrils collected in ultracentrifuged sediment fractions, the amyloid molecules produced from APPs include oligomeric non-fibrous assemblies in addition to soluble monomers. "A ⁇ oligomers” as described herein refer to non-fibrous assemblies. The “A ⁇ oligomers” as described herein include, for example, A ⁇ 40 (A ⁇ 1-40) oligomers and A ⁇ 42 (A ⁇ 1-42) oligomers.
  • a ⁇ 42 oligomers as described herein are molecules showing a molecular weight of 45 to 160 kDa in SDS-PAGE, and 22.5 to 1,035 kDa in Blue Native PAGE. Using molecular sieves, the molecules are collected mainly in the >100 kDa retention solution. When observed under an atomic force microscope, the molecules show mixed morphologies of granular, bead-shaped, and ring-shaped molecules having a height of 1.5 to 3.1 nm. By the gel filtration method, the molecules were eluted in the void volume fraction 8 with a molecular weight of 680 kDa or more, and in fraction 15 with a molecular weight of 17 to 44 kDa.
  • Antibodies of the present invention are monoclonal antibodies.
  • the antibodies of the present invention also include any type of antibodies such as non-human animal antibodies, humanized antibodies, chimeric antibodies, human antibodies, the later-described minibodies, amino acid sequence-modified antibodies, modified antibodies conjugated to other molecules (for example, polymers such as polyethylene glycol), and sugar chain-modified antibodies.
  • monoclonal antibodies refers to antibodies that are obtained from a substantially homogeneous population of antibodies. That is, the individual antibodies constituting the population are identical with the exception of possible natural mutants that may be present in a trace amount. Monoclonal antibodies are highly specific and recognize a single antigenic site. Each of the monoclonal antibodies recognizes a single determinant of the antigen, in contrast to conventional (polyclonal) antibody preparations that typically contain different antibodies against different antigenic determinants (epitopes).
  • monoclonal antibodies have the advantage that they can be synthesized from a hybridoma culture that is not contaminated with other immunoglobulins. Therefore, “monoclonal” indicates the characteristics of antibodies that can be obtained from a substantially homogeneous antibody population. This term does not indicate the requirement for any specific method for antibody production.
  • monoclonal antibodies can be produced by using known techniques. For example, they may be produced by the hybridoma method first described by Kohler and Milstein (Nature 256: 495-7, 1975 ), or by the recombinant DNA method ( Cabilly et al., Proc. Natl. Acad. Sci. USA 81:3273-7, 1984 ), but the methods are not limited thereto.
  • an A ⁇ oligomer for example, the A ⁇ tetramer described in the Examples
  • immunization is carried out according to a conventional immunization method.
  • the obtained immune cells are fused with known parent cells by a conventional cell fusion method, and monoclonal antibody-producing cells can be screened and isolated using a conventional screening method.
  • the monoclonal antibodies of the present invention can be produced as follows. Synthetic A ⁇ 1-42 (Peptide Institute, Inc., Osaka) is dissolved in distilled deionized water or a 10 mM phosphate buffer solution, and this is incubated at 37°C for 18 hours. Then, the peptides are separated by 4-12% SDS-PAGE, and visualized by CBB staining, and the portion of the A ⁇ 1-42 tetramer alone which is not contaminated with the A ⁇ 1-42 monomer is cut out and used as an antigen.
  • a preparation containing a large amount of the A ⁇ 1-40 oligomer is prepared by mixing (i) a modified A ⁇ 1-40 prepared by chemically linking 6-carboxytetramethylrhodamine (6-TAMRA) (SIGMA) to the N terminus of a synthetic A ⁇ 1-40 peptide using a conventional method with (ii) synthetic A ⁇ 1-40 (Peptide Institute, Inc., Osaka) at a ratio of 5:100, 10:100, 20:100, 30:100, 40:100, 50:100, 60:100, 70:100, or 80:100, preferably 90:100, or more preferably 100:100, and carrying out polymerization reaction for three hours, preferably six hours, or more preferably 20 hours.
  • 6-TAMRA 6-carboxytetramethylrhodamine
  • mice are immunized with 2.5 ⁇ g of either the A ⁇ 1-42 tetramer or A ⁇ 1-40 oligomer emulsified using complete Freund's adjuvant by injecting the antigen into their foot pad. Subsequently, booster immunizations are carried out six times. Hybridomas are produced from the inguinal lymph node by fusion with Sp2/O-Ag14 cells using Polyethylene Glycol 1500.
  • the animals immunized with sensitizing antigens are not particularly limited, but are preferably selected considering the compatibility with parent cells used for cell fusion.
  • rodents, lagomorphs, or primates are used.
  • Rodents include, for example, mice, rats, and hamsters.
  • Lagomorphs include, for example, rabbits.
  • Primates include, for example, Catarrhini (old-world) monkeys such as Macaca fascicularis, Macaca mulatta, hamadryas, and chimpanzees.
  • Animals are immunized with sensitizing antigens according to known methods. For example, as a standard method, immunization is performed by intraperitoneal or subcutaneous injection of a sensitizing antigen into mammals.
  • Cell fusion between the aforementioned immunocyte and a myeloma cell can be carried out basically according to known methods including the method by Kohler and Milstein ( Kohler G. and Milstein C., Methods Enzymol. (1981) 73, 3-46 ).
  • Hybridomas obtained in this manner are selected by culturing them in a conventional selection culture medium such as a HAT culture medium, which contains hypoxanthine, aminopterin, and thymidine. Culturing in the above-mentioned HAT culture medium is generally continued for several days to several weeks for an adequate time for killing cells other than the desired hybridomas (non-fused cells). Next, a conventional limiting dilution method is performed for screening and singly-cloning of a hybridoma that produces the desired antibody.
  • a conventional selection culture medium such as a HAT culture medium, which contains hypoxanthine, aminopterin, and thymidine. Culturing in the above-mentioned HAT culture medium is generally continued for several days to several weeks for an adequate time for killing cells other than the desired hybridomas (non-fused cells).
  • a conventional limiting dilution method is performed for screening and singly-cloning of a hybridoma that produces the desired
  • the obtained hybridoma is transplanted into the abdominal cavity of a mouse, and ascitic fluid containing the desired monoclonal antibodies is extracted.
  • the antibodies can be purified from the ascitic fluid by conventional protein separation and/or purification methods such as a selected combination of column chromatography including, but not limited to, affinity chromatography, filtration, ultrafiltration, salt precipitation, dialysis, SDS polyacrylamide gel electrophoresis, and isoelectric focusing ( Antibodies: A Laboratory manual, Harlow and David, Lane (edit.), Cold Spring Harbor Laboratory, 1988 ).
  • Protein A columns and Protein G columns can be used for affinity columns.
  • Examples of the Protein A columns used include Hyper D, POROS, and Sepharose F.F. (Pharmacia).
  • Chromatography includes ion exchange chromatography, hydrophobic chromatography, gel filtration, reverse-phase chromatography, and adsorption chromatography (" strategies for Protein Purification and Characterization: A Laboratory Course Manual", Daniel R Marshak et al., Cold Spring Harbor Laboratory Press, 1996 ).
  • liquid-phase chromatography methods such as HPLC and FPLC can be used.
  • Monoclonal antibody-producing hybridomas prepared in this manner can be subcultured in a conventional culture medium, and they can be stored for a long time in liquid nitrogen.
  • Any mammal can be immunized using an immunogen for antibody production.
  • the compatibility with parent cells used in cell fusion for hybridoma production is preferably considered.
  • rodents are used for the immunization.
  • Rodents include, for example, mice, rats, and hamsters.
  • Lagomorphs include, for example, rabbits.
  • Primates include, for example, Catarrhini (old-world) monkeys such as Macaca fascicularis, Macaca mulatta, hamadryas, and chimpanzees.
  • Catarrhini old-world monkeys
  • the use of transgenic animals that have a human antibody gene repertoire is known in the art ( Ishida I, et al., Cloning and Stem Cells 4: 91-102, 2002 ).
  • the transgenic animals are immunized, then antibody-producing cells are collected from the animals and fused with myeloma cells to produce hybridomas, and anti-protein human antibodies can be prepared from these hybridomas (see International Publication Nos. WO92/03918 , WO94/02602 , WO94/25585 , WO96/33735 , and WO96/34096 ).
  • lymphocytes immortalized with oncogenes may be used for monoclonal antibody production.
  • human lymphocytes infected with EB virus or such is immunized in vitro with immunogens.
  • the immunized lymphocytes are fused with human-derived myeloma cells (U266, etc) capable of unlimited division, and thus hybridomas that produce the desired human antibodies are obtained (Japanese Patent Application Kokai Publication No. ( JP-A) S63-17688 (unexamined, published Japanese patent application)).
  • the antibodies may also be prepared using genetic engineering methods (see, for example, Borrebaeck CAK and Larrick JW, Therapeutic Monoclonal Antibodies, MacMillan Publishers, UK, 1990 ).
  • recombinant antibodies may be prepared by cloning DNAs that encode the desired antibodies from antigen-producing cells such as hybridomas or immunized lymphocytes that produce the antibodies, then inserting the cloned DNAs into appropriate vectors, and transfecting the vectors into suitable host cells.
  • antigen-producing cells such as hybridomas or immunized lymphocytes that produce the antibodies
  • the monoclonal antibodies of the present invention include the 4H5 monoclonal antibody.
  • the monoclonal antibodies of the invention include an antibody that binds to an A ⁇ oligomer but not to an A ⁇ monomer, wherein the antibody is a monoclonal antibody and is an antibody that comprises an H chain (heavy chain) having the amino acid sequence of SEQ ID NO: 61 and an L chain (light chain) having the amino acid sequence of SEQ ID NO: 63.
  • the antibodies of the present invention include minibodies.
  • a minibody contains an antibody fragment lacking a portion of a whole antibody, and is not particularly limited as long as it has the ability to bind to an antigen.
  • antibody fragments include Fab, Fab', F(ab')2, and Fv.
  • minibodies include Fab, Fab', F(ab')2, Fv, scFv (single chain Fv), diabody, and sc(Fv)2 (single chain (Fv)2).
  • blood is removed from a mammal sensitized with an antigen after the serum level of the desired antibody is confirmed to be increased.
  • Serum is separated from blood by a known method.
  • serum containing the polyclonal antibody may be utilized.
  • a fraction containing the polyclonal antibody may be isolated from serum and then used.
  • immunoglobulin G or M can be prepared by obtaining a fraction that specifically recognizes a protein disclosed herein using an affinity column coupled with the protein, and then purifying this fraction using a Protein A or Protein G column.
  • an antibody that binds to an A ⁇ oligomer which is an antibody binding to an A ⁇ oligomer that binds 1A9, 2C3, 5A5, 5A9, 4F7, 4H5, 6E4, or 6H4.
  • the disclosed antibody is any one of the antibodies of (A) to (F) below:
  • a ⁇ oligomers are disclosed to which the antibodies bind.
  • the antibodies include, for example, the 4H5 monoclonal antibody.
  • Such A ⁇ oligomers can be used as antigens for preparing antibodies, or vaccines.
  • the antibodies disclosed include, for example, the antibody of any one of (1) to (38) below:
  • VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 9 (sequence of the 5A5 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 11 (sequence of the 5A5 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 13 (sequence of the 5A5 antibody H-chain CDR3) as CDR3"of (1) is a VH having the amino acid sequence of SEQ ID NO: 5 (sequence of the 5A5 antibody VH).
  • VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 15 (sequence of the 5A5 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 17 (sequence of the 5A5 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 19 (sequence of the 5A5 antibody L-chain CDR3) as CDR3"of (2) is a VL having the amino acid sequence of SEQ ID NO: 7 (sequence of the 5A5 antibody VL).
  • VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 29 (sequence of the 5A9 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 31 (sequence of the 5A9 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 33 (sequence of the 5A9 antibody H-chain CDR3) as CDR3"of (7) is a VH having the amino acid sequence of SEQ ID NO: 25 (sequence of the 5A9 antibody VH).
  • VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 49 (sequence of the 4F7 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 51 (sequence of the 4F7 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 53 (sequence of the 4F7 antibody H-chain CDR3) as CDR3"of (13) is a VH having the amino acid sequence of SEQ ID NO: 45 (sequence of the 4F7 antibody VH).
  • VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 55 (sequence of the 4F7 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 57 (sequence of the 4F7 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 59 (sequence of the 4F7 antibody L-chain CDR3) as CDR3"of (14) is a VL having the amino acid sequence of SEQ ID NO: 47 (sequence of the 4F7 antibody VL).
  • VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 89 (sequence of the 6E4 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 91 (sequence of the 6E4 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 93 (sequence of the 6E4 antibody H-chain CDR3) as CDR3"of (25) is a VH having the amino acid sequence of SEQ ID NO: 85 (sequence of the 6E4 antibody VH).
  • VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 95 (sequence of the 6E4 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 97 (sequence of the 6E4 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 99 (sequence of the 6E4 antibody L-chain CDR3) as CDR3"of (26) is a VL having the amino acid sequence of SEQ ID NO: 87 (sequence of the 6E4 antibody VL).
  • VH in the above-mentioned "H chain having the amino acid sequence of SEQ ID NO: 109 (sequence of the 6H4 antibody H-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 111 (sequence of the 6H4 antibody H-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 113 (sequence of the 6H4 antibody H-chain CDR3) as CDR3"of (31) is a VH having the amino acid sequence of SEQ ID NO: 105 (sequence of the 6H4 antibody VH).
  • VL in the above-mentioned "L chain having the amino acid sequence of SEQ ID NO: 115 (sequence of the 6H4 antibody L-chain CDR1) as CDR1, the amino acid sequence of SEQ ID NO: 117 (sequence of the 6H4 antibody L-chain CDR2) as CDR2, and the amino acid sequence of SEQ ID NO: 119 (sequence of the 6H4 antibody L-chain CDR3) as CDR3"of (32) is a VL having the amino acid sequence of SEQ ID NO: 107 (sequence of the 6H4 antibody VL).
  • the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 1 and SEQ ID NO: 2, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 3 and SEQ ID NO: 4, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 5 and SEQ ID NO: 6, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 7 and SEQ ID NO: 8, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 9 and SEQ ID NO: 10, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 11 and SEQ ID NO: 12, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown
  • the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 21 and SEQ ID NO: 22, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 23 and SEQ ID NO: 24, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 25 and SEQ ID NO: 26, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 27 and SEQ ID NO: 28, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 29 and SEQ ID NO: 30, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 31 and SEQ ID NO: 32, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are
  • the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 41 and SEQ ID NO: 42, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 43 and SEQ ID NO: 44, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 45 and SEQ ID NO: 46, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 47 and SEQ ID NO: 48, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 49 and SEQ ID NO: 50, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 51 and SEQ ID NO: 52, respectively; the amino acid sequence and the nucleo
  • the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 61 and SEQ ID NO: 62, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 63 and SEQ ID NO: 64, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 65 and SEQ ID NO: 66, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 67 and SEQ ID NO: 68, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 69 and SEQ ID NO: 70, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 71 and SEQ ID NO: 72,
  • the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 81 and SEQ ID NO: 82, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 83 and SEQ ID NO: 84, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 85 and SEQ ID NO: 86, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 87 and SEQ ID NO: 88, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 89 and SEQ ID NO: 90, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 91 and SEQ ID NO: 92, respectively;
  • the amino acid sequence and the nucleotide sequence of the full-length H chain are shown in SEQ ID NO: 101 and SEQ ID NO: 102, respectively; the amino acid sequence and the nucleotide sequence of the full-length L chain are shown in SEQ ID NO: 103 and SEQ ID NO: 104, respectively; the amino acid sequence and the nucleotide sequence of the H-chain variable region (VH) are shown in SEQ ID NO: 105 and SEQ ID NO: 106, respectively; the amino acid sequence and the nucleotide sequence of the L-chain variable region (VL) are shown in SEQ ID NO: 107 and SEQ ID NO: 108, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR1 are shown in SEQ ID NO: 109 and SEQ ID NO: 110, respectively; the amino acid sequence and the nucleotide sequence of the H-chain CDR2 are shown in SEQ ID NO: 111 and SEQ ID NO: 112, respectively;
  • the above-mentioned antibodies of (1) to (38) including the antibodies of the present invention include not only monovalent antibodies but also multivalent antibodies with two or more valencies.
  • the multivalent antibodies of the present invention include multivalent antibodies whose antigen binding sites are all the same and multivalent antibodies whose antigen binding sites are partially or completely different.
  • the above-mentioned antibody of (37) is an antibody with no modified CDRs.
  • the "antibody that comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of (1), which has equivalent activity as the antibody of (1)" of the above-mentioned antibody of (37) is preferably "an antibody that has equivalent activity as the antibody of (1), and comprises one or more amino acid substitutions, deletions, additions, and/or insertions in the antibody of (1), and comprises an H chain having the amino acid sequence of SEQ ID NO: 9 as CDR1, the amino acid sequence of SEQ ID NO: 11 as CDR2, and the amino acid sequence of SEQ ID NO: 13 as CDR3".
  • Another antibody of the above-mentioned antibody of (37) can be expressed in a similar manner.
  • equivalent activity means that the antibody of interest has biological or biochemical activity similar to that of an antibody of the present disclosure.
  • activities include the activity to bind specifically to A ⁇ oligomers but not to A ⁇ monomers, anti-neurotoxic activity, activity to suppress A ⁇ amyloid fibril formation, anti-synaptic toxicity activity, and anti-memory impairment activity.
  • Methods for preparing a polypeptide having activity equivalent to that of a certain polypeptide include methods for introducing mutations into a polypeptide.
  • one skilled in the art can prepare an antibody having activity equivalent to that of an antibody of the present invention by introducing appropriate mutations into the antibody using site-directed mutagenesis ( Hashimoto-Gotoh, T. et al. (1995) Gene 152, 271-275 ; Zoller, MJ, and Smith, M. (1983) Methods Enzymol. 100, 468-500 ; Kramer, W. et al. (1984) Nucleic Acids Res. 12, 9441-9456 ; Kramer W, and Fritz HJ (1987) Methods. Enzymol.
  • the antibodies of the present disclosure also include an antibody that comprises an amino acid sequence with one or more amino acid mutations in the amino acid sequence of an antibody of the present invention, and which has activity equivalent to that of the antibody of the present invention.
  • the number of mutated amino acids in such mutants may be generally 50 amino acids or less, preferably 30 amino acids or less, and more preferably ten amino acids or less (for example, five amino acids or less).
  • amino acids are categorized as follows depending on the side chain properties: hydrophobic amino acids (A, I, L, M, F, P, W, Y, and V), hydrophilic amino acids (R, D, N, C, E, Q, G, H, K, S, and T), amino acids with aliphatic side chains (G, A, V, L, I, and P), amino acids with hydroxyl-containing side chains (S, T, and Y), amino acids with sulfur atom-containing side chains (C and M), amino acids with carboxylic acid- and amide-containing side chains (D, N, E, and Q), amino acids with basic side chains (R, K, and H), and amino acids with aromatic ring-containing side chains (H, F, Y, and W) (amino acids are represented by one-letter codes in parentheses).
  • a polypeptide having an amino acid sequence, in which one or more amino acid residues are modified (deleted, added, and/or substituted with other amino acids) in a certain amino acid sequence, is known to retain its original biological activity (function).
  • the antibodies of the present invention may be conjugated to other substances as long as the activity is maintained.
  • the substances include peptides, lipids, sugars and sugar chains, acetyl groups, and natural and synthetic polymers. These modifications may be performed to confer additional functions to the antibodies, or to stabilize the antibodies.
  • Antibodies in which several amino acid residues have been added to the amino acid sequence of an antibody of the present invention include fusion proteins containing the antibody. In the fusion proteins, the antibody is fused with another peptide or protein. Methods for producing a fusion protein can be carried out by ligating a polynucleotide encoding an antibody of the present invention in frame with a polynucleotide encoding another peptide or polypeptide, and inserting this into an expression vector, and expressing the fusion construct in a host. Techniques known to those skilled in the art can be used for this purpose.
  • the peptides or polypeptides fused with an antibody of the present invention include, for example, known peptides such as FLAG ( Hopp, T.P.
  • 6x His consisting of six histidine (His) residues, 10x His, Influenza hemagglutinin (HA), human c-myc fragments, VSV-GP fragments, p18HIV fragments, T7-tag, HSV-tag, E-tag, SV40T antigen fragments, Ick tag, ⁇ -tubulin fragments, B-tag, and Protein C fragments; glutathione-S-transferase (GST); immunoglobulin constant regions; ⁇ -galactosidase; and maltose-binding protein (MBP), etc.
  • His histidine
  • HA Influenza hemagglutinin
  • HSV-GP fragments Influenza hemagglutinin
  • p18HIV fragments T7-tag
  • HSV-tag HSV-tag
  • E-tag E-tag
  • SV40T antigen fragments Ick tag
  • ⁇ -tubulin fragments B-tag
  • Protein C fragments Protein C
  • polynucleotides encoding these peptides or polypeptides can be fused with polynucleotides encoding the antibodies of the present invention, and the fusion polypeptides can be produced by expressing the fusion polynucleotides thus prepared.
  • the antibodies of the present invention may differ in the amino acid sequence, molecular weight, presence or absence of sugar chains, structure and such, depending on the cell or host producing the antibodies or the purification method.
  • Antibodies that bind to an epitope to which an antibody of any one of (1) to (36) above binds can be obtained by methods known to those skilled in the art.
  • the antibodies can be obtained by (i) determining the epitope bound by the antibody of any one of (1) to (36) using a conventional method, and producing the antibodies using a polypeptide comprising an amino acid sequence included in the epitope as an immunogen; or (ii) determining the epitopes of antibodies produced by a conventional method, and selecting antibodies whose epitope is the same as that of the antibody of any one of (1) to (36).
  • the above-mentioned antibodies of (1) to (38) also include any type of antibodies such as the above-described minibodies, antibodies with modified amino acid sequences such as humanized antibodies and chimeric antibodies, non-human animal antibodies, human antibodies, modified antibodies conjugated to other molecules (for example, polymers such as polyethylene glycol), and sugar chain-modified antibodies.
  • antibodies with modified amino acid sequences such as humanized antibodies and chimeric antibodies, non-human animal antibodies, human antibodies, modified antibodies conjugated to other molecules (for example, polymers such as polyethylene glycol), and sugar chain-modified antibodies.
  • the antibodies of the present invention are modified antibodies such as chimeric antibodies and humanized antibodies.
  • modified antibodies include (i) a chimeric antibody whose variable region is derived from the 4H5 antibody, and whose constant region is derived from a human immunoglobulin; and (ii) a humanized antibody whose CDR is derived from the 4H5 antibody, and whose FR is derived from a human immunoglobulin, and whose constant region is derived from a human immunoglobulin.
  • modified antibodies can be produced using known methods.
  • Chimeric antibodies are produced by combining sequences derived from different animals.
  • Examples of chimeric antibodies include antibodies comprising the heavy-chain and light-chain variable regions of a mouse antibody and the heavy-chain and light-chain constant regions of a human antibody.
  • the production of chimeric antibodies can be carried out using known methods (see, for example, Jones et al., Nature 321:522-5, 1986 ; Riechmann et al., Nature 332:323-7, 1988 ; and Presta, Curr. Opin. Struct. Biol. 2:593-6, 1992 ).
  • genes encoding the variable regions or CDRs of the antibody of interest are prepared from the RNAs of antibody-producing cells by polymerase chain reaction (PCR) or such (see, for example, Larrick et al., "Methods: a Companion to Methods in Enzymology", Vol. 2: 106, 1991 ; Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies” in Monoclonal Antibodies: Production, Engineering and Clinical Application; Ritter et al.
  • PCR polymerase chain reaction
  • the prepared genes encoding the variable regions are linked to genes encoding the constant regions or framework regions.
  • the genes encoding the constant regions or framework regions may be determined in a manner similar to that for the CDR-encoding genes, or alternatively, they can be prepared based on the sequence information of known antibodies.
  • DNA sequences encoding chimeric products and CDR-grafted products may be synthesized completely or partially using oligonucleotide synthesis techniques.
  • oligonucleotide synthesis described by Jones et al. (Nature 321:522-5, 1986 ) may be performed.
  • site-directed mutagenesis and polymerase chain reaction techniques may be appropriately used.
  • Techniques for oligonucleotide-specific mutagenesis of cases, site-directed mutagenesis and polymerase chain reaction techniques may be appropriately used.
  • CDR-grafting techniques are known in the art (" Immunoglobulin genes", Academic Press (London), pp 260-74, 1989 ; and Michael A et al., Proc. Natl. Acad. Sci. USA 91: 969-73, 1994 ).
  • the CDRs of a certain antibody are replaced with the CDRs of another antibody.
  • the binding specificity of the former antibody is changed to that of the latter antibody.
  • chimeric antibodies those in which the framework amino acids are derived from a human antibody are called "humanized antibodies (CDR-grafted antibodies)".
  • human antibodies or humanized antibodies are preferably utilized.
  • chimeric antibodies comprise the variable regions of a non-human mammal-derived antibody and the constant regions derived from a human antibody.
  • humanized antibodies comprise the complementarity-determining regions of a non-human mammal-derived antibody and the framework regions and constant regions derived from a human antibody.
  • amino acids in the variable regions for example, FRs
  • constant regions may be substituted with other amino acids.
  • the origin of the variable regions of the chimeric antibodies or the CDRs of the humanized antibodies is not particularly limited.
  • Human antibody-derived C-regions are used for the C-regions of the chimeric antibodies and humanized antibodies.
  • C ⁇ 1, C ⁇ 2, C ⁇ 3, C ⁇ 4, C ⁇ , C ⁇ , C ⁇ 1, C ⁇ 2, and C ⁇ can be used for the H-chain C-regions
  • C ⁇ and C ⁇ can be used for the L-chain C-regions.
  • Their sequences are known.
  • the human antibody C regions can be modified to improve the stability of the antibodies or their production.
  • the binding activity of the antibodies of the present invention to the antigens (A ⁇ oligomers) can be measured using, for example, an absorbance measurement method, an enzyme-linked immunosorbent assay (ELISA) method, an enzyme immunoassay (EIA) method, a radioimmunoassay (RIA) method, and/or a fluoroimmunoassay method.
  • ELISA enzyme-linked immunosorbent assay
  • EIA enzyme immunoassay
  • RIA radioimmunoassay
  • fluoroimmunoassay method e.g., an antibody is immobilized on a plate, and an antigen for the antibody is added to the plate, then a sample containing the desired antibody, such as the culture supernatant of antibody-producing cells or a purified antibody is added.
  • a secondary antibody which recognizes the primary antibody and is tagged with an enzyme such as alkaline phosphatase is added to the plate, and this is preincubated.
  • an enzyme substrate such as p -nitrophenyl phosphate is added to the plate, and the absorbance is measured to evaluate the antigen-binding ability of the sample of interest.
  • the evaluation can be performed using BIAcore (Pharmacia).
  • compositions comprising the above-mentioned antibody of the present invention and a pharmaceutically acceptable carrier.
  • the present invention strongly suggests that monoclonal 1A9 and 2C3 antibody are promising candidates for therapeutic antibodies for preventing Alzheimer-like phenotypes.
  • Memory deterioration has been shown to be related to synaptic dysfunction caused by soluble A ⁇ oligomers ( Klein WL, 2001, Trends Neurosci ; and Selkoe DJ, 2002, Science ).
  • Excessive accumulation and deposition of A ⁇ oligomers may trigger the complicated downstream cascades that cause Alzheimer's disease. If this is the case, therapeutic intervention using a composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier could be effective for blocking the pathologic cascades, and thus this could enable the treatment of Alzheimer's disease.
  • treatment of the present invention does not necessarily have complete therapeutic or preventive effects against organs or tissues exhibiting symptoms of disorders or diseases, but may have partial effects.
  • “Treatment of Alzheimer's disease” in the present invention means amelioration of at least one symptom that may be caused by Alzheimer's disease, and examples include amelioration or suppression of cognitive impairment, amelioration or suppression of senile plaque formation, amelioration or suppression of synaptic dysfunction, and reduction or suppression of A ⁇ accumulation in brain tissues, blood, or such.
  • cognitive impairment includes, for example, memory impairment including long term/short term memory impairment, object recognition memory impairment, spatial memory impairment, and associative and emotional memory impairment.
  • the present invention provides pharmaceutical compositions or pharmaceutical agents which comprise as an active ingredient the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier.
  • the phrase "comprising as an active ingredient the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier” means comprising the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier as a major ingredient, but does not limit its content rate.
  • compositions include agents against cognitive impairment, Alzheimer's disease agents, agents for suppressing the progression of Alzheimer's disease, agents for suppressing senile plaque formation, agents for suppressing A ⁇ accumulation, anti-neurotoxic agents (agents for neutralizing neurotoxicity), agents for inhibiting A ⁇ amyloid fibril formation, and anti-synaptic toxicity agents (agents for neutralizing synaptic toxicity).
  • the above-mentioned pharmaceutical composition of the present invention can be used, for example, in methods for suppressing Alzheimer's disease which comprise the step of administering to a subject (individual) the above-described composition comprising an antibody of the present invention and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition of the present invention for use include methods for suppressing cognitive impairment, methods for suppressing the progression of Alzheimer's disease, methods for suppressing senile plaque formation, methods for suppressing A ⁇ accumulation, methods for neutralizing (suppressing) neurotoxic activity, methods for inhibiting A ⁇ amyloid fibril formation, and methods for neutralizing (suppressing) synaptic toxicity.
  • Further examples of a pharmaceutical composition of the present invention for use include the use in methods for preventing and/or treating cognitive impairment, and methods for preventing and/or treating Alzheimer's disease.
  • the present invention also provides use of a composition comprising the above-described antibody of the present invention and a pharmaceutically acceptable carrier in the production of the above-mentioned pharmaceutical composition.
  • the present invention relates to the following compositions.
  • non-human animals to which the pharmaceutical agents are administered include mice, rats, guinea pigs, rabbits, chickens, cats, dogs, sheep, pigs, cattle, monkeys, baboons, and chimpanzees. These animals preferably exhibit at least one symptom selected from, for example, cognitive impairment, senile plaque formation, synaptic dysfunction, A ⁇ accumulation in brain tissues or blood, etc.
  • Antibodies contained in the pharmaceutical compositions of the present invention are not particularly limited as long as they are included in the above-mentioned antibodies of the present invention, and examples include the antibodies described herein.
  • the antibodies of the present invention may be formulated by methods known to those skilled in the art.
  • they can be prepared in the form of injectable sterile solutions or suspensions using water or another pharmaceutically acceptable liquid, and can be administered parenterally.
  • the antibodies to be included in the pharmaceutical compositions can be combined with acceptable carriers or media, specifically, sterile water, physiological saline, vegetable oils, emulsifiers, suspensions, surfactants, stabilizers, flavoring agents, excipients, solvents, preservatives, binders, or such, and mixed into a unit dose form required for generally accepted pharmaceutical practice.
  • phrases "pharmaceutically acceptable” indicates that the substance is inactive, and contains conventional substances used as diluents or vehicles for pharmaceuticals. Suitable excipients and their formulations are described, for example, in Remington's Pharmaceutical Sciences, 16th ed. (1980) Mack Publishing Co., ed. Oslo et al.
  • Physiological saline and other isotonic solutions containing glucose or adjuvants can be used as aqueous solutions for injection. They can be used together with appropriate solubilizers such as alcohols, more specifically, ethanol and polyalcohols (propylene glycol, polyethylene glycol, and such), and non-ionic surfactants (Polysorbate 80TM, HCO-50, and such).
  • solubilizers such as alcohols, more specifically, ethanol and polyalcohols (propylene glycol, polyethylene glycol, and such), and non-ionic surfactants (Polysorbate 80TM, HCO-50, and such).
  • Sesame oil or soybean oil can be used as an oleaginous liquid, and benzyl benzoate or benzyl alcohol can be used in combination as a solubilizer.
  • Buffers for example, phosphate buffer and sodium acetate buffer
  • soothing agents for example, procaine hydrochloride
  • stabilizers for example, benzyl alcohol and phenol
  • antioxidants can be used for the formulations.
  • Prepared injection solutions can be filled into appropriate ampules.
  • the administration is preferably parenteral administration, and specific examples include administration by injection, transnasal administration, transpulmonary administration, and transdermal administration.
  • administration by injection include systemic and local administration by intravenous injection, intramuscular injection, intraperitoneal injection, subcutaneous injection, and such.
  • compositions contain a pharmaceutically effective amount of the active component (the above-mentioned antibody of the present invention).
  • “Pharmaceutically effective amount (of a compound)” refers to an amount sufficient for treating and/or preventing disorders in which the antigens for the above-mentioned antibodies of the present invention play an important role.
  • a pharmaceutically acceptable amount may be an amount required for reducing A ⁇ accumulation, neutralizing A ⁇ -induced toxicity, reducing A ⁇ fibril formation, or such, thereby treating or preventing conditions caused by Alzheimer's disease, when the compound is administered to individuals (patients).
  • the reduction or neutralization may be, for example, a reduction or neutralization of at least approximately 5%, 10%, 20%, 30%, 40%, 50%, 75%, 80%, 90%, 95%, 99%, or 100%.
  • a suitable administration method may be selected depending on the age and symptoms of the patient.
  • the dosage of an antibody-containing pharmaceutical composition may be selected, for example, within the range of 0.0001 mg to 1000 mg per kilogram body weight for each administration.
  • the dosage for each patient may be selected within the range of 0.001 to 100,000 mg/body; however, the dosage is not necessarily limited to these ranges.
  • the dosage and administration methods vary depending on the patient's body weight, age, symptoms, and such, one skilled in the art can appropriately select them. In the later-described animal experiments, the dosage was selected based on the high-dose intravenous immunoglobulin therapy (400 mg/kg) covered by health insurance for humans.
  • the present invention provides methods for detecting A ⁇ oligomers (examples include A ⁇ 40 (A ⁇ 1-40) and A ⁇ 42 (A ⁇ 1-42) oligomers) in samples.
  • samples include samples collected from subjects.
  • the present methods include the step of detecting A ⁇ oligomers contained in a sample collected from a subject using an antibody of the present invention.
  • a ⁇ oligomers in a sample can be detected using, for example, sandwich solid-phase enzyme immunoassay methods that use chemiluminescence (chemiluminescence ELISA), immunoprecipitation methods that use the obtained antibodies, immunoblotting, flow cytometry, mass spectrometry, and immunohistochemical analysis.
  • sandwich solid-phase enzyme immunoassay methods that use chemiluminescence (chemiluminescence ELISA), immunoprecipitation methods that use the obtained antibodies, immunoblotting, flow cytometry, mass spectrometry, and immunohistochemical analysis.
  • the subject may be an Alzheimer's disease patient.
  • the amount of A ⁇ oligomers in a sample collected from a subject is compared with that from a healthy individual, and if the amount of A ⁇ oligomers is greater in the subject than in the healthy individual, the subject is determined to be a possible Alzheimer's disease patient.
  • a subject is a possible Alzheimer's disease patient is diagnosed usually by physicians (including individuals under instructions from physicians; same herein below).
  • Data on the amount of A ⁇ oligomers in samples collected from a subject and a healthy individual, which are obtained by the present methods of diagnosis, will be useful for diagnosis by physicians. Therefore, the present methods of diagnosis can be expressed as methods of collecting and presenting data useful for diagnosis by physicians.
  • the present invention provides methods for diagnosing whether or not a subject is a possible Alzheimer's disease patient, wherein the methods comprise detecting A ⁇ oligomers in a sample collected from the subject using an antibody of the present invention.
  • the present invention provides methods of diagnosing whether or not a subject is a possible Alzheimer's disease patient, which comprise the steps of:
  • a sample collected from a subject is contacted with an antibody of the present invention and an antibody that binds to an A ⁇ monomer.
  • contact may be carried out, for example, by adding each of the above-mentioned antibodies to a sample collected from a subject, which is placed in a test tube.
  • the antibody is added suitably in the form of a solution, a solid obtained by freeze-drying, or such.
  • the solution may purely contain the antibody alone, or may contain, for example, surfactants, excipients, coloring agents, flavors, preservatives, stabilizers, buffers, suspending agents, tonicity agents, binding agents, disintegrants, lubricants, fluidity promoters, or corrigents.
  • concentration at which the antibody is added is not particularly limited. For example, as with human immunoglobulin formulations, 500-mg, 1000-mg, and 2500-mg freeze-dried formulations and such may be suitably used.
  • the ratio of A ⁇ oligomer to A ⁇ monomer (herein, this is also referred to as "O/M index") in the aforementioned sample is measured.
  • the following method is suitably used. For example, as described below in the Examples, the measurement can be carried out using a method of comparing the oligomer and monomer ELISA values obtained from the same sample.
  • this ratio is compared with the ratio for a healthy individual.
  • the subject is determined to be a possible Alzheimer's disease patient.
  • the methods of diagnosis of the present invention can be performed both in vitro and in vivo, but they are preferably performed in vitro.
  • the "sample” described herein is not particularly limited as long as it is a tissue derived from a subject.
  • examples include the brain (brain parenchyma, and such), organs, and body fluids (blood, cerebrospinal fluid, and such) of a subject.
  • the sample is blood (more preferably, plasma) or cerebrospinal fluid.
  • the present invention provides pharmaceutical agents for use in the above-mentioned methods of measuring A ⁇ oligomers in a sample, or methods of diagnosing whether or not a subject is a possible Alzheimer's disease patient.
  • the pharmaceutical compositions comprising an antibody may be included in products and kits containing materials useful for treating pathological conditions of a subject.
  • the products may comprise any labeled container for a compound. Suitable containers include bottles, vials, and test tubes.
  • the containers may be formed from a variety of materials such as glass and plastic.
  • the label on the container surface should indicate that the composition is used to treat or prevent one or more conditions of the disease.
  • the label may also indicate descriptions for administration, and such.
  • kits containing a pharmaceutical composition comprising an antibody may optionally include a second container that stores a pharmaceutically acceptable diluent.
  • the kit may further include other materials desirable from a commercial and user's standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with descriptions for use.
  • the pharmaceutical compositions may be provided in a pack or dispenser device that may contain one or more unit dosage forms comprising an active ingredient.
  • the pack may comprise metal or plastic foil, and, for example, it is a blister pack.
  • the pack or dispenser device may be accompanied by instructions for administration.
  • the present inventors showed that the antibodies of the present invention are effective for preventing Alzheimer's disease. That is, the present invention provides methods for suppressing the progression of Alzheimer's disease, wherein the methods comprise the step of administering to an individual affected with Alzheimer's disease, a composition comprising the above-mentioned antibody of the present invention and a pharmaceutically acceptable carrier.
  • Synthetic A ⁇ 1-42 (Peptide Institute, Inc., Osaka) was dissolved in distilled water or 10 mM phosphate buffer, and incubated at 37°C for 18 hours. Then, the peptides were separated by SDS-PAGE (4-12% NuPAGE Tris-Glycine gel), and after visualization by CBB staining, just the A ⁇ 1-42 tetramer was excised without contamination of the A ⁇ 1-42 monomer.
  • a fluorescent dye, 6-carboxytetramethylrhodamine (6-TAMRA) (SIGMA) was chemically linked to the N terminus of a synthetic A ⁇ 1-40 peptide (Peptide Institute, Inc.) to produce a modified A ⁇ .
  • An oligomer-rich sample (A ⁇ 1-40 oligomer) was prepared by copolymerizing the modified A ⁇ and synthetic A ⁇ 1-40 peptide. It is preferable to adjust the conditions so that the fluorescence intensity determined by ThT assay, which is described below, is one-fourth or less the fluorescence intensity in the absence of modified A ⁇ . More specifically, it is preferred that 100 ⁇ M each of the modified A ⁇ and synthetic A ⁇ 1-40 peptide are mixed, and polymerized for 20 hours.
  • mice were immunized by injecting the antigen prepared by the method described above into their foot pads. Then, booster immunization was carried out six times. Hybridomas were prepared from inguinal lymph nodes by fusion with Sp2/O-Ag14 cells using Polyethylene Glycol 1500.
  • Isotyping of purified immunoglobulins was carried out using a Serotec (Oxford, UK) mouse monoclonal antibody isotyping kit.
  • the initial screening was carried out by dot blot analysis using a nitrocellulose membrane onto which 2.5 ⁇ l of A ⁇ 1-42 (2.5 ⁇ g/dot) pre-incubated for 18 hours was immobilized. Non-specific binding sites on the membrane were blocked with a phosphate buffer containing 5% low-fat milk, 1% BSA, and 0.05% Tween-20, and then the membrane was incubated with a culture supernatant.
  • a ⁇ oligomer-binding antibodies in the culture supernatant were detected by horseradish peroxidase-labeled goat anti-mouse F(ab') 2 (1:3000; Amersham), and visualized using an enhanced chemiluminescence (ECL) kit and LAS3000 mini (Fujitsu, Tokyo, Japan).
  • ECL enhanced chemiluminescence
  • the immunoprecipitated A ⁇ oligomers were separated using an NuPAGE 4-12% Bis-Tris-Glycine gel, and transferred onto a nitrocellulose membrane or Immobilon P (Millipore) using 10 mM 3-cyclohexylamino-1-propane sulfonic acid (pH 11) containing 10% methanol at 400 mA for one hour. Non-specific binding sites on the membrane were blocked with a phosphate buffer containing 5% low-fat milk, 1 % BSA, and 0.05% Tween-20 at room temperature for three hours.
  • the immunoprecipitated A ⁇ oligomers were detected by immunoblotting using the 4G8 (1:1000) or 6E10 (1:1000) monoclonal antibody as described above. Two clones, 1A9 and 2C3, were selected from the 16 clones as candidates for therapeutic antibodies for Alzheimer's disease.
  • the 6E10 and 4G8 monoclonal antibodies recognize the epitopes at amino acid positions 1-16 and 17-24 of the human A ⁇ sequence, respectively.
  • Polyclonal A11 which specifically recognizes A ⁇ oligomers was purchased from Biosource (Camarillo, CA).
  • Alex Fluor(AF)488- or 594-conjugated goat anti-mouse IgG and Alex Fluor(AF)488-conjugated goat anti-rat IgG were purchased from Molecular Probes (Eugene, OR).
  • Anti-mouse IgG2b was purchased from Sigma (St. Louis, MO).
  • An anti-synaptophysin antibody was purchased from Santa Cruz (Santa Cruz, CA), and an anti-drebrin antibody was purchased from MBL (Nagoya, Japan).
  • this concentrate was fractionated into 28 one-milliliter fractions using a Superose 12 size exclusion column (1 cm x 30 cm; Pharmacia Biotech., Uppsala, Sweden; flow rate of 0.5 ml/min) pre-equilibrated with a phosphate buffer.
  • Half of each fraction was subjected to immunoprecipitation using 1A9 or 2C3.
  • a ⁇ contained in the resulting immunoprecipitates was detected by immunoblotting using 4G8.
  • Synthetic A ⁇ 1-42 was dissolved at 250 ⁇ M in 0.02% ammonia water. Then, in order to prepare a seed-free A ⁇ solution, insoluble peptides, which may function as a seed, were precipitated by ultracentrifugation using an Optima TL ultracentrifuge (Beckman, USA) at 540,000 x g for three hours. The resulting supernatant was collected, aliquoted, and stored at -80°C until use. Samples were prepared by thawing the A ⁇ stock solutions immediately before use, and diluting them ten-fold with Tris-buffered saline (TBS; 150 mM NaCl and 10 mM Tris-HCl (pH 7.4)). The resulting 25 ⁇ M solutions were used in the experiments described below. Synthetic A ⁇ 1-40 (HCL form; Peptide Institute, Inc., Osaka) was prepared at 2x concentration.
  • An A ⁇ solution (25 ⁇ M) was incubated in the presence of a predetermined concentration of an antibody at 37°C for two or 24 hours.
  • the ThT fluorescence intensity of the incubation mixture was determined using a fluorescence spectrophotometer (RF-5300PC; Shimadzu Co., Kyoto, Japan).
  • the optimal fluorescence intensity was determined for A ⁇ amyloid fibrils at excitation and emission wavelengths of 446 and 490 nm, respectively, using 1.0 ml of a reaction mixture containing 5 ⁇ M ThT and 50 mM glycine-NaOH (pH 8.5). The fluorescence intensity was determined immediately after preparation of the mixture.
  • the activity of the 4F7, 4H5, 5A5, 5A9, 6E4, and 6H4 antibodies to suppress A ⁇ amyloid fibril formation was assessed by the following procedure.
  • An A ⁇ 1-42 solution diluted to 12.5 ⁇ M with cell culture medium was incubated in the presence or absence of each antibody at 37°C for 24 hours.
  • the amount of formed amyloid fibrils was determined by the above-described ThT fluorescence intensity assay method.
  • Rat pheochromocytoma PC12 cells were cultured in Dulbecco's Modified Eagle Medium (DMEM) (Invitrogen, Carlsbad, CA) containing 10% heat-inactivated horse serum (Invitrogen) and 5% fetal bovine serum (FBS) (Invitrogen).
  • DMEM Dulbecco's Modified Eagle Medium
  • FBS fetal bovine serum
  • PC12 cells were plated at a density of 20,000 cells/cm 2 in culture dishes coated with poly-L-lysine (10 mg/ml), and cultured for six days in DMEM supplemented with 100 ng/ml nerve growth factor (NGF; Alornone Labs, Jerusalem, Israel) (PC12N).
  • NGF nerve growth factor
  • PC12N was exposed to 25 ⁇ M seed-free A ⁇ 1-42 or pre-incubated A ⁇ 1-42 in the presence or absence of antibody at 4°C for 48 hours.
  • the neurotoxicity induced by A ⁇ 1-42 was assessed by Live/Dead dual-color fluorescence assay according to the supplier's instructions (Molecular Probes, Eugene, Oregon).
  • the four types of filtrates ( ⁇ 3 kDa, 3 to 10 kDa, 10 to 30 kDa, 30 to 100 kDa) and the retention solution (>100 kDa) were prepared from a 25 ⁇ M A ⁇ oligomer solution by sequential ultrafiltration using Microcon 3 kDa, 10 kDa, 30 kDa, and 100 kDa cut-off membranes.
  • Each of the fractions was subjected to the A ⁇ -induced neurotoxicity assay described above.
  • PC12N was exposed to each fraction to identify the toxic fraction as described above.
  • the distribution of the three-dimensional structures recognized by A11, 1A9, 2C3, and 4G8 was also identified by the dot blot analysis described above.
  • the morphological characterization of the neurotoxic oligomers was performed by examining each fraction using an atomic force microscope.
  • Electron microscopy (EM) and atomic force microscopy (AFM) are Electron microscopy (EM) and atomic force microscopy (AFM)
  • Samples were diluted with distilled water and sprayed over carbon-coated grids to conduct electron microscopy.
  • the grids were negatively-stained with 1% phosphotungstic acid and observed under a Hitachi H-7000 electron microscope (Tokyo, Japan) with an acceleration voltage of 77 kV.
  • AFM assessment was carried out as recently reported. Drops of the samples were placed onto freshly cleaved mica. The mica was allowed to stand for 30 minutes and then washed with water, and the liquid samples were analyzed using Nanoscope IIIa (Digital Instruments, Santa Barbara, CA, USA) set to the tapping mode ( Tero, R, et al., Langmuir 20, 7526-7531, 2004 ).
  • the cantilever used was OMCL-TR400PSA (Olympus, Japan).
  • Frozen tissue samples (the anterior portion of entorhinal cortex) were homogenized in nine volumes of Tris-buffered saline (TS) containing a protease inhibitor cocktail. The homogenates were ultracentrifuged at 265,000 x g for 20 minutes. One-third aliquots (0.5 ml) of the resulting supernatants were subjected to immunoblot analysis.
  • TS Tris-buffered saline
  • the left brain hemispheres of Tg2576 mice were sliced into 30- ⁇ m-thick sagittal sections using a cryotome (RM 2145; Leica, Wetzlar, Germany), and stained with thioflavin S as previously described (Wyss-Coray et al., 2001).
  • the formation of swollen dystrophic neurites was observed using an anti-synaptophysin antibody (Chemicon, Temecula, CA).
  • the number of thioflavin S-positive plaques and synaptophysin-positive swollen dystrophic neurites was counted by observing four or five sections from the left brain hemisphere of each mouse at 40-fold magnification.
  • a ⁇ deposition serial sections briefly pre-treated with formic acid or Protease K were stained using an A ⁇ immunostaining kit (Sigma, St. Louis, MO), and immuno-positive signals were visualized using an ABC elite kit (Vector Laboratories). Images of the cerebral cortex and hippocampus were recorded using a digital camera connected with a microscope, and analyzed using a simple PCI software (Compix Imaging System, Lake Oswego, OR). The brain translocation of antibodies was observed using a confocal laser microscope (Carl Zeiss LSM510). The number of thioflavin S-positive plaques and synaptophysin-positive swollen dystrophic neurites was determined in a double blind manner.
  • mice Three-month-old female non-transgenic (non-Tg) mice, and Tg2576 mice having and overexpressing the Swedish-type mutant human APP gene with dual mutations (K670N and M671L) derived from familial AD were purchased from Taconics (Germantown, NY, USA). These mice were reared until 13 months old in the animal facility of the present inventors.
  • 1A9 or 2C3 0.4 mg/kg/week
  • PBS was administered into the caudal vein of four-month-old Tg2576, and the administration was continued until 13 months.
  • the memory function was assessed at month 13 as previously described ( Mouri A, FASEB J, 21: 2135-2148, 2007 ), based on the following four behavioral paradigms:
  • CSF was collected from 12 AD patients and 13 NC individuals. Then, lipoproteins were removed from 600 ⁇ l each of the CSF by preparative continuous density gradient ultracentrifugation according to a protocol reported previously ( Matsubara E, et al., Ann Neurol, 45: 537-541, 1999 ). The density of CSF was adjusted to 1.25 g/ml with KBr. The CSF was ultracentrifuged at 100,000 rpm and 16°C for eight hours using a Hitachi RP100AT centrifuge.
  • Lipoproteins floating at a density of 1.25 g/ml and lipoprotein-depleted CSF (LPD-CSF) were subjected to ultrafiltration using a 3 kDa cut-off membrane (Microcon 3; Arnicon, Inc), and then frozen and stored, or stored at 4°C, until use.
  • LPD-CSF lipoprotein-depleted CSF
  • Lipoproteins were also removed by affinity chromatography using PHML-LIPOSORB (Calbiochem, La Jolla, CA). Each sample (plasma or brain) and PHML-LIPOSORB (Calbiochem, La Jolla, CA) were combined at a ratio of 1.5:1, and mixed for 60 seconds. Then, the mixture was centrifuged at 3,000 rpm for ten minutes. The resulting supernatants (lipoprotein-free samples) were subjected to ELISA using 6E10 for the oligomers. The lipoprotein-bound samples were eluted from PHML-LIPOSORB using 20 mM sodium deoxycholate. The removal of specific lipoproteins was confirmed by agarose electrophoresis using 1% gel (Beckmann), followed by staining with FAST-RED 7B (Wako, Osaka, Japan).
  • Chemiluminescence-based sandwich solid-phase enzyme immunoassay was used to specifically detect oligomeric A ⁇ but not monomeric A ⁇ .
  • Microplates were coated with monoclonal 1A9 (IgG2b isotype) or 2C3 (IgG2b isotype), or a mixture of 1A9 and 2C3. 100 ⁇ l of a sample (brain or cerebrospinal fluid) was added and incubated continuously for 24 hours at 4°C.
  • horseradish peroxidase-conjugated BA27 Fab' fragment (anti-A ⁇ 1-40 specific to A ⁇ 40; Wako pure chemical, Osaka, Japan) or horseradish peroxidase-conjugated BCO5 Fab' fragment (anti-A ⁇ 35-43 specific to A ⁇ 42; Wako pure chemical, Osaka, Japan) was added and incubated at 4°C for 24 hours.
  • the chemiluminescence generated using SuperSignal ELISA Pico Chemiluminescent Substrate (Pierce, Rockford, IL, USA) was quantified by a Veritas Microplate Luminometer (Promega).
  • a ⁇ oligomers used in this assay were prepared by diluting synthetic A ⁇ 1-40 (HCl form) to a concentration of 0.1 mg/ml with PBS and incubating this at 37°C for one hour. Meanwhile, A ⁇ monomers were prepared by diluting synthetic A ⁇ 1-40 (TFA form) to a concentration of 0.1 mg/ml with PBS. A ⁇ oligomers were immobilized onto 96-well immunoplates at 400 ng/well, and then the plates were blocked with BSA.
  • the A ⁇ monomers or oligomers stepwise-diluted in the range of 100 pg/ml to 100 ⁇ g/ml were reacted with the 4F7, 4H5, 5A5, 5A9, 6E4, or 6H4 antibodies, or the control anti-A ⁇ antibodies (4G8 and 6E10). After incubation for two hours, the mixtures were added to the above-described 96-well immunoplates, and incubated at room temperature for ten minutes. The binding of immobilized A ⁇ oligomers to each of the antibodies was detected by measuring the absorbance at 450 nm in the color development reaction using an HRP-labeled anti-mouse IgG antibody and a TMB solution.
  • a ⁇ oligomers and monomers co-exist in a solution. Thus, it is essential to remove A ⁇ monomers for preparation of antigens to produce A ⁇ oligomer-specific antibodies.
  • the present inventors succeeded in isolating SDS-stable A ⁇ tetramers without contamination of A ⁇ monomers by SDS-PAGE.
  • a phosphate buffer-insoluble and formic acid (FA)-soluble amyloid fraction derived from AD brain was analyzed by immunoprecipitation using the cell culture supernatants of the positive hybridomas ( Fig. 1B ).
  • the A ⁇ dimer, a smaller amount of the trimer, and a high-molecular-weight smear characteristic to aggregated A ⁇ molecular species were detected by immunoblot analysis using anti-A ⁇ monoclonal 4G8. A very small amount of A ⁇ monomers dissociated in the presence of SDS was also detected.
  • oligomers To further confirm the existence of three-dimensional structures recognized by native 1A9 and 2C3 (i.e ., oligomers), the present inventors detected the oligomers in conditioned medium (CM) of human embryonic kidney (HEK) 293 cells transfected with mutant PS1 cDNA ( Nakaya Y et al., J Biol Chem, 280: 19070-19077, 2005 ). The present inventors fractionated HEK293 CM by SEC, and then identified the oligomers.
  • CM conditioned medium
  • HEK 293 cells transfected with mutant PS1 cDNA
  • NGF-differentiated PC12 cells PC12N
  • 25 ⁇ M seed-free A ⁇ 1-42 ThiT-negative 540,000 x g supernatant
  • mAbs monoclonal antibodies
  • the viability of nerve cells was determined by LIVE/DEAD assay ( Fig. 2 ). Nerve cell death was detected at a significantly high level (50%) in the presence of A ⁇ 1-42 ( Figs. 2B and 2G ), as compared to the control assay ( Fig. 2A ).
  • Non-specific IgG2b Figs.
  • each fraction was subjected to atomic force microscopy (AFM).
  • AFM atomic force microscopy
  • the presence of globular particle morphology consistent with the fraction size was detected in the three neurotoxic fractions.
  • Fig. 3E shows the atomic force microscopic images of non-toxic fraction 2 (Fr. 2), toxic fractions 3 (Fr. 3) and 4 (Fr. 4), and the most toxic fraction 5 (Fr. 5).
  • the formation of many granular polymer molecules was clearly observed in the toxic fractions.
  • fraction 5 was revealed to contain heterogeneous toxic molecules including bead-shaped and ring-shaped molecules in addition to various large and small granular molecules.
  • a ⁇ 1-42 amyloid fibrils at 0, 10, 25, and 50 ⁇ M was assessed by measuring the ThT fluorescence for 72 hours at 37°C.
  • seed-free A ⁇ 1-42 ThiT-negative supernatant fraction obtained by ultracentrifugation at 540,000 x g was polymerized into amyloid fibrils by nucleation-dependent polymerization ( Fig. 4A ).
  • the present inventors incubated 25 ⁇ M seed-free A ⁇ 1-42 in the presence or absence of the antibodies at 37°C for 48 hours. As shown in Fig. 4B , the ThT fluorescence intensity was altered in a 2C3 concentration-dependent manner, while none of monoclonal 1A9 and 4G8 and non-specific IgG2b altered the florescence intensity. Meanwhile, when A ⁇ was polymerized by incubation for two hours, 1A9, as well as 2C3, showed the activity to almost completely suppress the fibril formation ( Fig. 4C ).
  • the polymerization time course was analyzed by dot blotting using A11, 1A9, 2C3, and 4G8.
  • the majority of A11 antibody-reactive oligomers was formed during the lag time phase of polymerization (0 to 8 hours), and the ThT fluorescence intensity was relatively weak.
  • the level of A11-immunoreactive oligomers reached a plateau, and then was constant (about 20% of the peak level) until 72 hours (plateau phase).
  • the 1A9-recognized oligomers were detected only after four hours, and then the immunoreactivity to 1A9 increased twofold over time. This suggests that the 1A9-recognized oligomers are slowly formed. Meanwhile, it was revealed that the 2C3-recognized oligomers are transiently increased during the lag time phase (0 to 8 hours), and then exist at a very low level (less than 5%) in a oligomerized state from 8 to 72 hours.
  • the HMW smear was dramatically reduced, and there were two types of major components: a high-molecular-weight species that could not enter the gel and thus remained in the well, and a small amount of the monomers.
  • the molecular sieve experiment revealed that seed-free A ⁇ 1-42 is converted into molecular species of 100 kDa or more, and exhibits the strongest toxicity.
  • the toxic molecules include molecular species showing a high-molecular-weight (HMW) smear pattern of 45 to 160 kDa, in addition to the monomer to pentamer species, and the toxic polymers can be easily depolymerized into low-molecular-weight species in the presence of SDS.
  • HMW high-molecular-weight
  • the neurotoxic activity of de novo -formed A ⁇ oligomers was reduced by about 12.5% and 26%, respectively ( Fig. 5C ).
  • the level of de novo -formed A ⁇ oligomers in the early period of A ⁇ polymerization is a determining factor for neurotoxicity, and that the formation reaches a peak in the period of zero to two hours, and then the level of formed A ⁇ oligomers reduces over time.
  • nuclei for the de novo polymerization of A ⁇ amyloid fibrils, or amyloid fibrils themselves have the neurotoxicity-neutralizing activity.
  • the present inventors incubated A ⁇ 1-42 for two hours, and then removed insoluble A ⁇ polymerization nuclei and amyloid fibrils by ultracentrifugation for three hours at 540,000 x g.
  • neurotoxic 1A9-recognized polymers are basically soluble toxic oligomers that have been slightly stabilized due to some structural change, while neurotoxic 2C3-recognized polymers are basically short-lived oligomeric intermediates that are very unstable due to drastic structural changes during the early stage of polymerization process.
  • Monoclonal 1A9 and 2C3 recognize A ⁇ oligomers in the brain parenchyma
  • the present inventors demonstrated the specificity and biological activity of 1A9 and 2C3. Furthermore, the inventors detected 1A9 and 2C3 polymers in the brain by immunohistochemistry.
  • the present inventors performed conventional immunohistochemistry methods to enhance immune reaction by formaldehyde fixation, and formic acid, SDS, or microwave treatment of brain sections. The two antibodies exhibited no immunoreactivity to AD brain by any one of the enhancement methods.
  • the present inventors pre-treated the sections with Protease K, which is known to improve immunostaining ( Wrzolek MA, et al., Am J Pathol, 141: 343-355, 1992 ).
  • Protease K which is known to improve immunostaining
  • Brain homogenates were prepared using Tris-buffered saline (TBS) to avoid chemical modification during the extraction of soluble oligomers.
  • TBS Tris-buffered saline
  • the oligomers having a molecular weight of 4mer, 5mer, 8mer, and 12mer were immunoprecipitated with 1A9 from TBS samples of the cerebral cortex from AD brain ( Fig. 6D , lane 2), while the level of the oligomers in the control healthy brain was below the detection limit (lane 3).
  • the level of 12mer in the buffer-soluble fractions of previously reported 50 autopsy cases was determined by immunoblotting using monoclonal 1A9 and 2C3.
  • the 50 cases include two AD cases, 35 cases at Braak NFT stages I to II, and 13 cases at NFT stages III to IV ( Katsuno et al., Neurology, 64: 687-692, 2005 ). As shown in Figs.
  • the A ⁇ polymers (soluble 1A9- and 2C3-imuunoreactive 12mer) responsible for the in vivo neurotoxicity were found in the brain parenchyma.
  • CSF also contains the polymers.
  • the BC05/BC05 oligomer ELISA detected soluble A ⁇ 1-42 in fraction 13
  • the BA27/BA27 ELISA detected soluble A ⁇ 1-40 in fractions 7 to 14 (data not shown).
  • the level of lipoprotein-bound A ⁇ monomer in AD was comparable to that of the healthy control, while the level of lipoprotein-free A ⁇ 40 monomer ( Fig. 7-1A ) and A ⁇ 42 monomer ( Fig. 7-1B ) in AD was lower as compared to the age-matched healthy control.
  • the present inventors also found that lipoprotein-bound A ⁇ monomers, in addition to the oligomers, can be detected when ELISA is designed to use HRP-labeled BC05 or BA27 as a capture antibody.
  • the present inventors improved the detection antibodies and samples used in ELISA.
  • Lipoproteins were pre-depleted from CSF, and the resulting lipoprotein-depleted CSF (LPD-CSF) was used as an assay sample.
  • a ⁇ oligomer-specific 1A9 and 2C3 were used as detection antibodies for ELISA.
  • chemiluminescence ELISA was developed to enhance the sensitivity. Pooled LPD-CSF ( Figs. 7-1C to D ) was fractionated by SEC, and each fraction was analyzed for A ⁇ oligomer distribution by luminescence ELISA using 1A9 or 2C3 as a detection antibody. As shown in Figs.
  • a ⁇ oligomers were detected in SEC fractions 12 to 15 (relatively large A ⁇ with a molecular weight ranging within 18 to 108 kDa, which corresponds to the size of 4mer to 24mer).
  • the level of 1A9- and 2C3-recognized oligomers was elevated in all of the AD patient-derived fractions in which the oligomers were detectable.
  • the level of A ⁇ oligomers in LPD-CSF from AD patients was compared to that from the age-matched healthy control, although a limited number of cases were analyzed. As shown in Fig.
  • the structural change from A ⁇ monomer to oligomer occurs in the earliest period of the process of A ⁇ polymerization.
  • the ratio between A ⁇ oligomer and monomer can be used as a clinical indicator reflecting the pathological conditions of AD.
  • O/M index can be used as a clinical indicator reflecting the pathological conditions of AD.
  • results described above show that the 1A9- and 2C3-positive three-dimensional structures are present as A ⁇ oligomers in LPD-CSF, and increased in AD patients.
  • results obtained by the present inventors demonstrated that the structural conversion of lipoprotein-free soluble A ⁇ to the oligomeric intermediates occurs in CSF of AD patients, and the oligomers can be detected as useful biological markers for diagnosis of sporadic AD.
  • PBS-administered Tg2576 mice showed significant learning and behavioral impairments ( Fig. 8A to 8D ).
  • the memory function of 1A9 and 2C3-administered Tg2576 mice was indistinguishable from that of age-matched non-administered wild type cohort mice, which was previously determined. Therefore, 1A9 and 2C3-administered Tg2576 mice were shown to retain both short- and long-term memory, which were impaired in the PBS administration group.
  • the present inventors obtained evidence supporting the view that the onset of memory disturbance, in particular AD, can be prevented by conducting passive immunotherapy targeting A ⁇ oligomers before the onset. Furthermore, the result described above presents the first in vivo evidence that directly indicates that A ⁇ oligomers are responsible for the onset of memory disturbance.
  • Monoclonal 1A9 prevents A ⁇ accumulation in the brain of Tg2576
  • the amount of A ⁇ accumulated in the brain was determined in the following three fractions (150 mg/extract) prepared by serial extraction: soluble fraction in Tris buffer containing protease inhibitors; 2% SDS-soluble amyloid fraction; and 2% SDS-insoluble and 70% formic acid-soluble amyloid fraction.
  • non-accumulative, physiological A ⁇ molecules are contained in the Tris buffer fraction, while 2% SDS-soluble A ⁇ includes A ⁇ in diffuse senile plaques before amyloid fibril formation, immunocytochemically undetectable A ⁇ , and conformationally altered, accumulative soluble oligomeric A ⁇ .
  • a ⁇ was selectively quantified by A ⁇ 40 and A ⁇ 42 end-specific ELISA (BNT77/BA27 specific for A ⁇ 40, BNT77/BC05 specific for A ⁇ 42, WAKO kit). There was no marked difference among the three groups in the A ⁇ concentration in the Tris buffer fraction where the major components were non-accumulative, physiological A ⁇ molecules ( Figs. 9A and 9C , A ⁇ x-40; Figs.
  • Plasma A ⁇ oligomers are increased by passive immunotherapy with 1A9 and 2C3
  • the present inventors assessed the level of physiological saline-soluble and -insoluble A ⁇ oligomers in pooled brain homogenates, and the level of A ⁇ oligomers in the peripheral blood and plasma. There was no difference in the amount of physiological saline-soluble A ⁇ oligomers in the pooled brain homogenates among the treatment groups ( Fig. 10D ). Meanwhile, the amount of insoluble A ⁇ oligomers was shown to be reduced in the 1A9 and 2C3 treatment groups ( Fig. 10E ).
  • pooled plasma from each group (albumin-depleted plasma, upper part of Panel F; albumin/lipoprotein-depleted plasma, lower part of Panel F) was assayed for A ⁇ oligomers by A11 dot blotting.
  • the result shows that the oligomers were present in the plasma from PBS-administered Tg2576 mice ( Fig. 10F ).
  • A11-positive oligomers in plasma were clearly increased in the passive immunotherapy groups as compared to the PBS administration group ( Fig. 10F ).
  • the proportion of 2C3-recognized oligomers in a lipoprotein-bound form was greater than that of 1A9-recognized oligomers (lower part of Panel F).
  • plasma A ⁇ oligomers were detected by A11 immunoprecipitation.
  • the result shows that the oligomers of about 200 kDa were increased in Tg2576 mice treated with passive immunotherapy as compared to the PBS administration group ( Fig. 10G ).
  • the increase in plasma A ⁇ oligomers in the passive immunotherapy groups can be considered to directly reflect enhanced cerebral clearance.
  • the present inventors obtained evidence that direct target molecules for intravenous passive immunotherapy are also present in blood in addition to brain, and that oligomer-selective cerebral clearance can be enhanced through peripheral sites of action. That is, the present inventors showed the clinical usefulness of the intravenous passive immunotherapy.
  • Fig. 11A Immunohistochemical A ⁇ deposition was suppressed in the passive immunotherapy groups.
  • Fig. 11B The formation of thioflavin S-positive senile amyloid plaques was significantly suppressed in both the cerebral cortex and hippocampus ( Fig. 11B , upper part), and the reduction was also clearly demonstrated by histochemistry ( Fig. 11B , lower part).
  • the formation of synaptophysin-positive swollen dystrophic neurites was also significantly suppressed in the passive immunotherapy groups ( Fig. 11C ).
  • the antibodies translocate to the brain
  • mice IgG The existence/localization of deposited A ⁇ and cerebral mouse IgG was assessed using a confocal laser microscope. The result shows that mouse IgG is localized almost independently of deposited A ⁇ within the areas containing diffuse senile plaques. Mouse IgG was observed only in the passive immunotherapy groups (1A9, Fig. 13A ; 2C3, Fig. 13B ), but not in the PBS administration group ( Fig. 13C ). Thus, a fraction of the antibodies administered into the blood was considered to translocate to the brain.
  • each antibody was mixed with stepwise-diluted A ⁇ oligomers or monomers ("inhibitors"), and the pre-mixed solutions were added to A ⁇ oligomer-immobilized 96-well immunoplates, and then incubated (see the "Methods" section).
  • the commercially available 4G8 and 6E10 antibodies were used as control antibodies that nonselectively bind to A ⁇ oligomers and monomers.
  • the antibody pre-mixed with A ⁇ monomers does not bind to the A ⁇ monomers in the solution, and therefore can bind to immobilized A ⁇ oligomers.
  • the antibody pre-mixed with A ⁇ oligomers binds to the A ⁇ oligomers in the solution, and therefore the amount of antibody bound to immobilized A ⁇ oligomers is reduced with the increase in inhibitor concentration.
  • the results for the six types of antibodies (4F7, 4H5, 5A5, 5A9, 6E4, and 6H4) showed concentration-dependent reduction in the amount of bound antibody when A ⁇ oligomers were used. In contrast, no such strong reduction in binding was detected when A ⁇ monomers were used (see Fig.
  • the remaining four types of antibodies (5A5, 5A9, 6E4, and 6H4) were found to markedly reduce the cytotoxicity (see Fig. 16 ).
  • the results described above demonstrate that the four types of antibodies (5A5, 5A9, 6E4, and 6H4) have a strong activity of neutralizing A ⁇ -induced neurotoxicity. Since 4F7 and 4H5 lowered the cytotoxicity as compared to control IgG, these antibodies are also inferred to have an activity of neutralizing A ⁇ -induced neurotoxicity.
  • the present inventors commenced treatment of young Tg2576 mice before the onset of memory disturbance with anti-A ⁇ oligomer passive immunotherapy targeting 1A9- and 2C3-recognized toxic polymers.
  • the present inventors presented evidence supporting that age-dependent memory deterioration that naturally develops in Tg2576 mice can be prevented by passive immunotherapy using anti-A ⁇ oligomer-specific antibodies (1A9 and 2C3).
  • short-term memory disturbance assessed by the Y-maze test is similar to the A ⁇ accumulation-associated memory disturbance observed in mild cognitive impairment (MCI) and early AD.
  • the Y-maze test showed excellent and almost normal results in Tg2576 mice administered with 1A9 and 2C3, respectively.
  • the novel object recognition task, Morris water maze, and contextual fear conditioning task the long-term memory was maintained nearly normal by the anti-A ⁇ oligomer antibodies.
  • a selective increase in A11-positive oligomers in blood was observed in the antibody-treated mouse groups as compared to the PBS treatment group, which is consistent with the ability of the antibodies to prevent the onset of memory disturbance (the memory maintenance ability).
  • the 1A9 antibody treatment also exhibited the effect of suppressing cerebral A ⁇ accumulation.
  • the 2C3 antibody treatment demonstrated a higher blood level of A11-positive oligomers as compared to the 1A9 antibody treatment.
  • the cerebral A ⁇ accumulation-suppressing effect of the 2C3 antibody treatment was unclear. Accordingly, 1A9-recognized oligomers were considered to have greater contribution to the cerebral A ⁇ accumulation than 2C3-recognized oligomers.
  • neurotoxic 1A9 polymers are soluble toxic oligomers that are somewhat conformationally, while neurotoxic 2C3 polymers are very unstable, short-lived oligomeric intermediates that appear at an early stage of the polymerization process, the conformation of which is easily changed.
  • the present inventors disclose herein the in vivo preventive effect of anti-oligomer antibodies on Alzheimer's disease, and this is the first evidence that directly demonstrates that toxic A ⁇ oligomers formed in vivo can inhibit the functions of nerve cells, thereby inducing the symptoms of Alzheimer's disease.
  • the data obtained by the present inventors is also the first evidence supporting the view that A ⁇ exhibits in vivo neurotoxicity in the human brain. It is well known that the human entorhinal cortex is an area that is easily affected with AD. In this area, NFT formation and nerve cell loss precede the formation of senile plaques. Thus, the entorhinal cortex is an exceptional area to which the commonly accepted amyloid cascade hypothesis cannot be applied. However, this inconsistency has been neglected and remained unstudied for a long time.
  • the present inventors proposed and examined the hypothesis that previously unidentifiable, invisible A ⁇ oligomers are harmful for nerve cells in the entorhinal cortex and cause memory disturbance.
  • the present inventors performed semi-quantitative analysis of 1A9- and 2C3-immunoreactive 12mer in the entorhinal cortex of elderly individuals who were mostly at Braak NFT stages I to III.
  • the 1A9- and 2C3-immunoreactive 12mer were already present in the entorhinal cortex of healthy individuals at Braak NFT stages I to II, and increased with the advancement of Braak NFT stage.
  • the 12mer was found to be significantly increased in AD.
  • the data of the present inventors show for the first time evidence that directly demonstrates in vivo the memory disturbance resulting from synaptic dysfunction caused by endogenous A ⁇ oligomers.
  • active immunotherapy Janus D, 2000, Nature ; Morgan D, 2000, Nature
  • passive immunotherapy Bard F, 2222, Nat med ; DeMattos RB, PNAS, 2001
  • the mechanism by which learning disability and memory disturbance can be prevented has remained a matter of conjecture.
  • One widely proposed possibility is that the antibodies reach the brain through the blood-brain barrier and directly neutralize in vivo soluble A ⁇ oligomers that cause memory impairment.
  • the second possibility, the "sink theory", is that the antibodies act peripherally to deplete the peripheral blood A ⁇ pool and thus activate A ⁇ clearance from the brain.
  • DeMattos et al. have reported that a peripherally administered anti-A ⁇ antibody rapidly transports not only cerebral A ⁇ monomers but also A ⁇ dimers into plasma, and also cerebral A ⁇ into CSF ( DeMattos RB et al., PNAS, 98; 8850-8855, 2001 ).
  • the present inventors also revealed that A ⁇ oligomers are present in human CSF and increased in AD patients. Thus, the present inventors demonstrated that the A ⁇ oligomers can be used as diagnostic markers for AD.
  • the present inventors presented the first evidence supporting the view that A ⁇ oligomers are present in the plasma of Tg2576 mice, and, in passive immunotherapy by which A ⁇ oligomers are specifically captured and neutralized through intravenous injection, intracerebral antibody delivery is not required and the clearance of A ⁇ oligomers from the brain to blood can be enhanced at the peripheral sites of action, i.e., blood vessels.
  • the present inventors presented the first evidence that passive immunotherapy can suppress senile amyloid plaque formation, and indirectly suppress nerve cell damage (swollen dystrophic neurite formation) through senile amyloid plaque suppression.
  • a ⁇ oligomer is the molecular basis for the onset of Alzheimer's disease, and selective control using oligomer-specific antibodies enables the control of Alzheimer's disease from a prophylactic viewpoint, in addition to a therapeutic viewpoint. Furthermore, a fraction of the administered antibodies was proven to translocate into the brain. This suggests that the effect of suppressing memory disturbance is exerted by a combination of multiple actions such as direct neutralization of soluble A ⁇ oligomers in the brain, transport of antibody-A ⁇ oligomer immune complexes into blood by the neonatal Fc receptor ( Deane R, 2005, J Neurosci ), and the "sink" action described above.
  • the antibodies provided by the present invention can be used, for example, in intravenous injection-based preventive passive immunotherapy for Alzheimer's disease, and as biological markers for pre-onset diagnosis, disease monitoring, drug efficacy monitoring/assessment for the disease, and such.
  • the antibodies of the present invention are expected to greatly contribute to the establishment of preventive/therapeutic methods for Alzheimer's disease that are selective to molecules responsible for evoking the pathological conditions of the disease, and the establishment of early diagnostic markers.
  • the present inventors obtained evidence supporting that antibody therapies, even when they target intracerebral pathological conditions, can be satisfactorily achieved by peripheral intravenous administration, without the need to consider intracerebral transfer of the antibodies.
  • the present inventors obtained evidence demonstrating that a fraction of administered antibodies translocates to the brain and produces a direct effect even in peripheral intravenous administration therapy, again without the need to consider intracerebral transfer of the antibodies.
  • the present invention is expected to rapidly accelerate the progress of antibody therapeutics for Alzheimer's disease.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Biomedical Technology (AREA)
  • Organic Chemistry (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Neurosurgery (AREA)
  • Neurology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Urology & Nephrology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Microbiology (AREA)
  • Pathology (AREA)
  • Cell Biology (AREA)
  • Biotechnology (AREA)
  • General Physics & Mathematics (AREA)
  • Analytical Chemistry (AREA)
  • Food Science & Technology (AREA)
  • Physics & Mathematics (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Dermatology (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Claims (15)

  1. Antikörper, der an ein Aβ-Oligomer bindet, aber nicht an einen Aß-Monomer, wobei der Antikörper ein monoklonaler Antikörper ist und ausgewählt ist aus der Gruppe bestehend aus:
    (1) ein Antikörper, der eine H-Kette, die die Aminosäuresequenz SEQ ID Nr: 69 als CDR1, die Aminosäuresequenz von SEQ ID Nr: 71 als CDR2 und die Aminosäuresequenz von SEQ ID Nr: 73 als CDR3 aufweist, und eine L-Kette, die die Aminosäuresequenz von SEQ ID Nr: 75 als CDR1, die Aminosäuresequenz von SEQ ID Nr: 77 als CDR2 und die Aminosäuresequenz von SEQ ID Nr: 79 als CDR3 aufweist, umfasst;
    (2) ein Antikörper, der eine H-Kette, die die Aminosäuresequenz von SEQ ID Nr: 65 als VH aufweist und eine L-Kette, die die Aminosäuresequenz von SEQ ID Nr: 67 als VL aufweist, umfasst; und
    (3) ein Antikörper, der eine H-Kette, die die Aminosäuresequenz von SEQ ID Nr: 61 aufweist, und eine L-Kette, die die Aminosäuresequenz von SEQ ID Nr: 63 aufweist, umfasst.
  2. Antikörper nach Anspruch 1, wobei der Antikörper ein chimärer Antikörper oder ein humanisierter Antikörper ist.
  3. Zusammensetzung umfassend den Antikörper nach Anspruch 1 oder 2, und einen pharmazeutisch annehmbaren Träger.
  4. Mittel zur Verwendung bei der Behandlung von kognitiver Beeinträchtigung, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  5. Therapeutisches Mittel zur Verwendung bei der Behandlung der Alzheimer-Erkrankung, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  6. Mittel zur Verwendung beim Unterdrücken des Fortschreitens von Alzheimer-Erkrankung, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  7. Mittel zur Verwendung beim Unterdrücken der Bildung seniler Plaques, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  8. Mittel zur Verwendung beim Unterdrücken der Aβ-Akkumulation, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  9. Mittel zur Verwendung beim Neutralisieren der Neurotoxizität, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  10. Mittel zur Verwendung beim Inhibieren der Bildung von Aβ-Amyloid Fibrillen, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  11. Mittel zur Verwendung beim Neutralisieren der synaptischen Toxizität, das den Antikörper nach Anspruch 1 oder 2 oder die Zusammensetzung nach Anspruch 3 als einen aktiven Wirkstoff umfasst.
  12. Verfahren zum Detektieren eines Aβ-Oligomers, das den Schritt des Detektierens eines Aβ-Oligomers umfasst, das in einer Probe enthalten ist, die einem Subjekt entnommen wurde, unter Verwendung des Antikörpers nach Anspruch 1 oder 2.
  13. Verfahren zum Diagnostizieren ob oder ob nicht ein Subjekt ein möglicher Alzheimer-Erkrankungs-Patient ist, welches umfasst
    den Schritt (a0) untenstehend:
    (a0) Verwenden des Antikörpers nach Anspruch 1 oder 2, um ein Aβ-Oligomer in einer Probe zu detektieren, die einem Subjekt entnommen wurde; oder
    die Schritte von (a1) bis (c1) untenstehend:
    (a1) Inkontaktbringen einer Probe, die einem Subjekt entnommen wurde, mit dem Antikörper nach Anspruch 1 oder 2;
    (b1) Messen der Menge an Aβ-Oligomer in der Probe; und
    (c1) Bestimmen, dass das Subjekt ein möglicher Alzheimer-Erkrankungs-Patient ist, falls die Menge, die in Schritt (b1) gemessen wird, höher ist als diejenige eines gesunden Individuums; oder
    die Schritte von (a2) bis (c2) untenstehend:
    (a2) Inkontaktbringen der Probe, die einem Subjekt entnommen wurde, mit dem Antikörper nach Anspruch 1 oder 2, und einem Antikörper, der an ein Aß-Monomer bindet;
    (b2) Messen des Verhältnisses von Aβ-Oligomer zu Aß-Monomer in der Probe; und
    (c2) Bestimmen, dass das Subjekt ein möglicher Alzheimer-Erkrankungs-Patient ist, falls das Verhältnis, das in Schritt (b2) gemessen wird, höher ist als dasjenige eines gesunden Individuums.
  14. Verfahren nach Anspruch 12 oder 13, wobei die Probe Blut oder Rückenmarkflüssigkeit ist.
  15. Pharmazeutisches Mittel zur Verwendung bei der Diagnose ob oder ob nicht ein Subjekt ein möglicher Alzheimer-Erkrankungs-Patient ist, das den Antikörper nach Anspruch 1 oder 2 umfasst.
EP16183829.7A 2008-02-08 2009-02-06 Zur spezifischen bindung an ab-oligomer fähiger antikörper und verwendung davon Active EP3121277B1 (de)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2008028386 2008-02-08
US8554508A 2008-08-01 2008-08-01
JP2008201058 2008-08-04
EP09707511.3A EP2246427B1 (de) 2008-02-08 2009-02-06 Antikörper mit fähigkeit zur spezifischen bindung an amyloid-beta oligomere und deren verwendung

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
EP09707511.3A Division EP2246427B1 (de) 2008-02-08 2009-02-06 Antikörper mit fähigkeit zur spezifischen bindung an amyloid-beta oligomere und deren verwendung
EP09707511.3A Division-Into EP2246427B1 (de) 2008-02-08 2009-02-06 Antikörper mit fähigkeit zur spezifischen bindung an amyloid-beta oligomere und deren verwendung

Publications (2)

Publication Number Publication Date
EP3121277A1 EP3121277A1 (de) 2017-01-25
EP3121277B1 true EP3121277B1 (de) 2018-04-11

Family

ID=40952248

Family Applications (2)

Application Number Title Priority Date Filing Date
EP09707511.3A Active EP2246427B1 (de) 2008-02-08 2009-02-06 Antikörper mit fähigkeit zur spezifischen bindung an amyloid-beta oligomere und deren verwendung
EP16183829.7A Active EP3121277B1 (de) 2008-02-08 2009-02-06 Zur spezifischen bindung an ab-oligomer fähiger antikörper und verwendung davon

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP09707511.3A Active EP2246427B1 (de) 2008-02-08 2009-02-06 Antikörper mit fähigkeit zur spezifischen bindung an amyloid-beta oligomere und deren verwendung

Country Status (15)

Country Link
US (3) US8378081B2 (de)
EP (2) EP2246427B1 (de)
JP (2) JP5113853B2 (de)
KR (1) KR101616136B1 (de)
CN (2) CN102936287B (de)
AU (2) AU2009211635B2 (de)
CA (1) CA2714413C (de)
DK (1) DK2246427T3 (de)
ES (1) ES2617604T3 (de)
HK (1) HK1181789A1 (de)
HU (1) HUE031944T2 (de)
PL (1) PL2246427T3 (de)
PT (1) PT2246427T (de)
SG (1) SG188116A1 (de)
WO (1) WO2009099176A1 (de)

Families Citing this family (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10277519B2 (en) * 2006-01-31 2019-04-30 Silicon Laboratories Inc. Response time for a gateway connecting a lower bandwidth network with a higher speed network
SI2436696T1 (sl) * 2007-01-05 2017-10-30 University Of Zurich Anti-beta-amiloidno protitelo in načini njegove uporabe
IL199534A (en) 2007-01-05 2013-01-31 Univ Zuerich An isolated human antibody capable of detecting a neoepitope in a disease-related protein, a polynucleotide encoding an antibody, a vector containing the polynucleotide, a host cell containing the polynucleotide or vector, a preparation containing the antibody and related methods and uses.
US7939075B2 (en) * 2007-01-11 2011-05-10 Philipps-Universitaet Marburg Human monoclonal anti-amyloid-beta antibodies
KR20100115340A (ko) * 2007-10-19 2010-10-27 이무나스 파마 가부시키가이샤 Aβ 올리고머에 특이적으로 결합하는 항체 및 그의 이용
US20100291071A1 (en) * 2008-08-01 2010-11-18 Immunas Pharma, Inc. Antibody Specific Binding to A-Beta Oligomer and the Use
CN102936287B (zh) * 2008-02-08 2015-09-09 伊缪纳斯制药株式会社 能够特异性结合Aβ寡聚体的抗体及其应用
CA2730073A1 (en) * 2008-07-09 2010-01-14 University Of Zurich Method of promoting neurogenesis
US9085614B2 (en) 2008-08-01 2015-07-21 Immunas Pharma, Inc. Antibodies that specifically bind to Aβ oligomers and uses thereof
JP5812418B2 (ja) 2009-04-17 2015-11-11 イムナス・ファーマ株式会社 Aβオリゴマーに特異的に結合する抗体およびその利用
ES2624835T3 (es) 2009-08-06 2017-07-17 Immunas Pharma, Inc. Anticuerpos que se unen específicamente a los oligómeros A beta y uso de los mismos
DK2462162T3 (en) * 2009-08-06 2017-01-16 Immunas Pharma Inc Antibodies that specifically bind to A-beta oligomers and their use
CN102597234B (zh) * 2009-08-07 2014-10-29 协和发酵麒麟株式会社 人源化抗淀粉样-b寡聚体抗体
EP2463369A4 (de) * 2009-08-07 2013-09-18 Kyowa Hakko Kirin Co Ltd Humanisierter antikörper gegen amyloid-b-oligomere
WO2011151076A2 (en) * 2010-06-04 2011-12-08 Georg-August-Universität Göttingen Stiftung Öffentlichen Rechts, Universitätsmedizin MONOCLONAL ANTIBODIES TARGETING Αβ OLIGOMERS
US9320793B2 (en) * 2010-07-14 2016-04-26 Acumen Pharmaceuticals, Inc. Method for treating a disease associated with soluble, oligomeric species of amyloid beta 1-42
WO2012012257A2 (en) * 2010-07-19 2012-01-26 Virginia Commonwealth University BIVALENT MULTIFUNCTIONAL LIGANDS TARGETING Aβ OLIGOMERS AS TREATMENT FOR ALZHEIMER'S DISEASE
US8785603B2 (en) * 2011-05-20 2014-07-22 Siemens Healthcare Diagnostics Inc. Antibodies to 25-hydroxyvitamin D2 and D3 and uses thereof
CA2867338A1 (en) * 2012-03-13 2013-09-19 Janssen Alzheimer Immunotherapy Oligomeric a.beta. in the diagnosis, prognosis, and monitoring of alzheimer's disease
WO2013167681A1 (en) 2012-05-10 2013-11-14 Georg-August-Universität Göttingen CONFORMATIONAL-SPECIFIC ANTIBODIES AGAINST Αβ OLIGOMERS
JP6092049B2 (ja) * 2013-08-28 2017-03-08 東芝ライフスタイル株式会社 撮像システム及び撮像装置
JP6582995B2 (ja) 2014-01-21 2019-10-02 株式会社島津製作所 App切断型ペプチドの測定方法
EP3783364A3 (de) 2014-05-22 2021-05-19 Shimadzu Corporation Surrogatbiomarker zur beurteilung der intrazerebralen amyloid-beta-peptid-akkumulation und verfahren zur analyse davon
EP3207057A2 (de) * 2014-10-16 2017-08-23 F. Hoffmann-La Roche AG Anti-alpha-synuklein-antikörper und verfahren zur verwendung
MA41115A (fr) 2014-12-02 2017-10-10 Biogen Int Neuroscience Gmbh Procédé de traitement de la maladie d'alzheimer
GB2541003A (en) * 2015-08-05 2017-02-08 Kran Life Sciences Llp Neurodegenerative disorders
EP3351939B1 (de) 2015-09-16 2020-11-04 Shimadzu Corporation Multiplex-biomarker zur verwendung in der beurteilung des zustands der anhäufung von amyloid b im gehirn und analyseverfahren für besagte beurteilung
JP6974348B2 (ja) 2016-01-09 2021-12-01 アーベル リミテッドArbele Limited がん処置のためのカドヘリン−17特異的抗体及び細胞傷害性細胞
JP2017132742A (ja) * 2016-01-29 2017-08-03 国立大学法人 大分大学 Aβオリゴマー特異的抗体の新規薬理用途
CN107118260B (zh) * 2017-05-12 2020-10-16 中国科学院过程工程研究所 一种多肽及其组成的疫苗和应用
SG11202001281WA (en) 2017-08-22 2020-03-30 Biogen Ma Inc Pharmaceutical compositions containing anti-beta amyloid antibodies
CA3114869C (en) 2018-10-01 2023-10-03 Toru Miyazaki Therapeutic agent for neurodegenerative disease
CN114269379A (zh) * 2019-06-28 2022-04-01 加利福尼亚大学董事会 用于治疗阿尔茨海默病的方法和组合物
JP7370569B2 (ja) * 2019-08-07 2023-10-30 国立大学法人 大分大学 アミロイドβタンパク質オリゴマーと結合するヒト化抗体
GB201912008D0 (en) * 2019-08-21 2019-10-02 Cambridge Entpr Ltd Antibody
US20220348645A1 (en) * 2019-10-01 2022-11-03 Beth Israel Deaconess Medical Center, Inc. Conformation-specific antibodies that bind nuclear factor kappa-light-chain-enhancer of activated b cells
CN116348487A (zh) 2020-07-23 2023-06-27 欧萨尔普罗席纳有限公司 抗淀粉样β抗体
CA3226568A1 (en) * 2021-07-19 2023-01-26 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Mct11 antibodies to treat t cell functional exhaustion and enhance cancer immunotherapy
AU2022386674A1 (en) * 2021-11-15 2024-05-30 Ascendo Biotechnology, Inc. Methods to reverse treml1-induced immune suppression
CN114605532B (zh) * 2022-05-07 2022-08-23 北京第一生物化学药业有限公司 抗β-淀粉样蛋白抗体及其用途
CN117088984B (zh) * 2022-05-11 2024-05-10 东莞市朋志生物科技有限公司 抗苯二氮卓抗体、检测苯二氮卓的试剂和试剂盒
CN116789818B (zh) * 2023-06-25 2024-01-26 东南大学 一种抗β淀粉样蛋白的单链抗体及应用

Family Cites Families (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP0239102A3 (de) 1986-03-28 1989-07-12 Tsuji, Kimiyoshi Verfahren zur Herstellung eines Human-Human-Hybridoms
IL162181A (en) 1988-12-28 2006-04-10 Pdl Biopharma Inc A method of producing humanized immunoglubulin, and polynucleotides encoding the same
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
WO1992003918A1 (en) 1990-08-29 1992-03-19 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
NZ255101A (en) 1992-07-24 1997-08-22 Cell Genesys Inc A yeast artificial chromosome (yac) vector containing an hprt minigene expressible in murine stem cells and genetically modified rodent therefor
EP0754225A4 (de) 1993-04-26 2001-01-31 Genpharm Int Heterologe antikörper produzierende transgene nicht-humane tiere
EP1978033A3 (de) 1995-04-27 2008-12-24 Amgen Fremont Inc. Aus immunisiertem Xenomid abgeleitete menschliche Antikörper
CA2219486A1 (en) 1995-04-28 1996-10-31 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6218506B1 (en) 1997-02-05 2001-04-17 Northwestern University Amyloid β protein (globular assembly and uses thereof)
US20030068316A1 (en) 1997-02-05 2003-04-10 Klein William L. Anti-ADDL antibodies and uses thereof
AR022952A1 (es) * 1999-03-19 2002-09-04 Smithkline Beecham Corp ANTICUERPO MONOCLONAL DE ROEDOR ESPECIFICAMENTE NEUTRALIZANTE PARA LA INTERLEUQUINA-18 HUMANA , UN FRAGMENTO FAB NEUTRALIZANTE o FRAGMENTO F(AB')2, UNA REGION DE COMPLEMENTARIEDAD DE CADENA LIGERA DE INMONOGLOBULINA(CDR), UNA MOLECULA DE ACIDO NUCLEICO, COMPOSICION FARMACEUTICA QUE LO COMPRENDE, EL
AUPR617901A0 (en) 2001-07-06 2001-08-02 Pacmab Pty Ltd Method for treating multiple myeloma
JP4729717B2 (ja) 2001-08-03 2011-07-20 株式会社医学生物学研究所 GM1ガングリオシド結合型アミロイドβタンパク質を認識する抗体、及び該抗体をコードするDNA
MY139983A (en) 2002-03-12 2009-11-30 Janssen Alzheimer Immunotherap Humanized antibodies that recognize beta amyloid peptide
WO2010012004A2 (en) 2008-07-25 2010-01-28 The Regents Of The University Of California Monoclonal antibodies specific for pathological amyoid aggregates common to amyloids formed from proteins of differing sequence
DE10303974A1 (de) 2003-01-31 2004-08-05 Abbott Gmbh & Co. Kg Amyloid-β(1-42)-Oligomere, Verfahren zu deren Herstellung und deren Verwendung
US8394379B2 (en) * 2003-05-15 2013-03-12 Iogenetics, Llc Targeted cryptosporidium biocides
JP2008513732A (ja) * 2004-07-02 2008-05-01 ノースウエスタン ユニバーシティ アミロイドβ(Abeta)の病理学的なアセンブリを標的とするモノクローナル抗体
US20080044356A1 (en) 2004-10-22 2008-02-21 Regents Of The University Of Minnesota Assemblies of Oligomeric Amyloid Beta Protein and Uses Thereof
AU2005306997B2 (en) * 2004-10-25 2012-07-05 Merck Sharp & Dohme Corp. Anti-ADDL antibodies and uses thereof
TW200635608A (en) * 2004-12-15 2006-10-16 Neuralab Ltd Aβ antibodies for use in improving cognition
AU2006211625A1 (en) 2005-01-14 2006-08-10 The Regents Of The University Of California Compositions and methods for inhibiting drusen formation and for diagnosing or treating drusen-related disorders
CA2589860A1 (en) 2005-01-24 2006-08-03 Amgen Inc. Humanized anti-amyloid antibody
US7731962B2 (en) 2005-02-14 2010-06-08 Merck & Co., Inc. Anti-ADDL monoclonal antibody and use thereof
EP1861422B1 (de) 2005-03-05 2010-02-24 Abbott GmbH & Co. KG Screening-verfahren, verfahren zur aufreinigung nichtdiffundierbarer a-beta-oligomere, selektive antikörper gegen diese nichtdiffundierbaren a-beta-oligomere und verfahren zur herstellung dieser antikörper
US7741448B2 (en) 2005-06-21 2010-06-22 Medical & Biological Laboratories Co., Ltd. Antibody having inhibitory effect on amyloid fibril formation
EP1749839A1 (de) 2005-07-22 2007-02-07 Novoplant GmbH Antigen-bindende Polypeptide gegen F4 (K88) fimbriae
CN101421303B (zh) * 2006-03-23 2013-06-12 生命北极神经科学公司 改进的初原纤维选择性抗体及其用途
US20090232801A1 (en) 2007-05-30 2009-09-17 Abbot Laboratories Humanized Antibodies Which Bind To AB (1-42) Globulomer And Uses Thereof
US20100291071A1 (en) 2008-08-01 2010-11-18 Immunas Pharma, Inc. Antibody Specific Binding to A-Beta Oligomer and the Use
KR20100115340A (ko) 2007-10-19 2010-10-27 이무나스 파마 가부시키가이샤 Aβ 올리고머에 특이적으로 결합하는 항체 및 그의 이용
CA2709354C (en) 2007-12-21 2014-06-17 Amgen Inc. Anti-amyloid antibodies and uses thereof
CN102936287B (zh) 2008-02-08 2015-09-09 伊缪纳斯制药株式会社 能够特异性结合Aβ寡聚体的抗体及其应用
US9085614B2 (en) 2008-08-01 2015-07-21 Immunas Pharma, Inc. Antibodies that specifically bind to Aβ oligomers and uses thereof
JP5812418B2 (ja) 2009-04-17 2015-11-11 イムナス・ファーマ株式会社 Aβオリゴマーに特異的に結合する抗体およびその利用
DK2462162T3 (en) 2009-08-06 2017-01-16 Immunas Pharma Inc Antibodies that specifically bind to A-beta oligomers and their use
ES2624835T3 (es) 2009-08-06 2017-07-17 Immunas Pharma, Inc. Anticuerpos que se unen específicamente a los oligómeros A beta y uso de los mismos

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
None *

Also Published As

Publication number Publication date
CA2714413C (en) 2017-01-24
ES2617604T3 (es) 2017-06-19
SG188116A1 (en) 2013-03-28
WO2009099176A1 (ja) 2009-08-13
JPWO2009099176A1 (ja) 2011-05-26
JP5113853B2 (ja) 2013-01-09
JP5765857B2 (ja) 2015-08-19
US20100028357A1 (en) 2010-02-04
EP3121277A1 (de) 2017-01-25
AU2009211635A1 (en) 2009-08-13
AU2009211635B2 (en) 2014-06-26
HUE031944T2 (en) 2017-08-28
US8378081B2 (en) 2013-02-19
US20110097319A1 (en) 2011-04-28
JP2013063976A (ja) 2013-04-11
DK2246427T3 (da) 2017-02-20
US20130149316A1 (en) 2013-06-13
EP2246427A4 (de) 2012-04-25
EP2246427A1 (de) 2010-11-03
PT2246427T (pt) 2017-03-03
EP2246427B1 (de) 2016-11-30
CA2714413A1 (en) 2009-08-13
KR20100113623A (ko) 2010-10-21
CN102936287B (zh) 2015-09-09
AU2014224074A1 (en) 2014-10-02
HK1181789A1 (en) 2013-11-15
KR101616136B1 (ko) 2016-04-27
CN102124105A (zh) 2011-07-13
PL2246427T3 (pl) 2017-06-30
CN102936287A (zh) 2013-02-20
US9090680B2 (en) 2015-07-28

Similar Documents

Publication Publication Date Title
EP3121277B1 (de) Zur spezifischen bindung an ab-oligomer fähiger antikörper und verwendung davon
EP2210901A1 (de) Zur spezifischen bindung an ein oligomer fähiger antikörper, und dessen verwendung
EP2462161B1 (de) Spezifisch an abeta-oligmomere bindende antikörper und ihre verwendung
EP2419447B1 (de) Spezifisch an abeta-oligmomere bindende antikörper und ihre verwendung
US20100291071A1 (en) Antibody Specific Binding to A-Beta Oligomer and the Use
US8613924B2 (en) Antibodies that specifically bind to A beta oligomers and use thereof
US9085614B2 (en) Antibodies that specifically bind to Aβ oligomers and uses thereof

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2246427

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20170404

RBV Designated contracting states (corrected)

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20171005

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AC Divisional application: reference to earlier application

Ref document number: 2246427

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 988056

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180415

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602009051813

Country of ref document: DE

REG Reference to a national code

Ref country code: ES

Ref legal event code: FG2A

Ref document number: 2670405

Country of ref document: ES

Kind code of ref document: T3

Effective date: 20180530

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20180411

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180711

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180711

Ref country code: SE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180712

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 988056

Country of ref document: AT

Kind code of ref document: T

Effective date: 20180411

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180813

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602009051813

Country of ref document: DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20190114

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190206

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20190228

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190206

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190228

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20190206

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180811

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20090206

REG Reference to a national code

Ref country code: DE

Ref legal event code: R082

Ref document number: 602009051813

Country of ref document: DE

Representative=s name: SIMMONS & SIMMONS LLP, DE

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20180411

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: FR

Payment date: 20230221

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20230223

Year of fee payment: 15

Ref country code: GB

Payment date: 20230221

Year of fee payment: 15

Ref country code: DE

Payment date: 20230216

Year of fee payment: 15

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: ES

Payment date: 20230427

Year of fee payment: 15

P01 Opt-out of the competence of the unified patent court (upc) registered

Effective date: 20230704